Bioluminescence Fundamentals and Applications in Biotechnology - 2014
Bioluminescence Fundamentals and Applications in Biotechnology - 2014
Bioluminescence Fundamentals and Applications in Biotechnology - 2014
Gérald Thouand
Robert Marks Editors
Bioluminescence:
Fundamentals and
Applications in
Biotechnology–
Volume 1
144
Advances in Biochemical
Engineering/Biotechnology
Series editor
T. Scheper, Hannover, Germany
Editorial Board
S. Belkin, Jerusalem, Israel
P. M. Doran, Hawthorn, Australia
I. Endo, Saitama, Japan
M. B. Gu, Seoul, Korea
S. Harald, Potsdam, Germany
W. S. Hu, Minneapolis, MN, USA
B. Mattiasson, Lund, Sweden
J. Nielsen, Gothenburg, Sweden
G. N. Stephanopoulos, Cambridge, MA, USA
R. Ulber, Kaiserslautern, Germany
A.-P. Zeng, Hamburg-Harburg, Germany
J.-J. Zhong, Shanghai, China
W. Zhou, Framingham, MA, USA
This book series reviews current trends in modern biotechnology and biochemical
engineering. Its aim is to cover all aspects of these interdisciplinary disciplines,
where knowledge, methods and expertise are required from chemistry, biochem-
istry, microbiology, molecular biology, chemical engineering and computer
science.
In general, volumes are edited by well-known guest editors. The series editor and
publisher will, however, always be pleased to receive suggestions and supple-
mentary information. Manuscripts are accepted in English.
Editors
Bioluminescence:
Fundamentals and
Applications in
Biotechnology–Volume 1
With contributions by
123
Editors
Gérald Thouand
UMR CNRS 6144
University of Nantes
La Roche-sur-Yon
France
Robert Marks
National Institute for Biotechnology
in the Negev
Ben Gurion University
Beersheba
Israel
and
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
v
Part I
Fundamentals of Bioluminescence
Eco-Evo Bioluminescence on Land
and in the Sea
Keywords Aposematism Coelenterazine Counter-illumination Cypridinid
luciferin DNA barcoding Ecology Evolution Symbiotic luminescence
Contents
1 Introduction.......................................................................................................................... 4
2 Function of Bioluminescence.............................................................................................. 4
2.1 Self-defense................................................................................................................. 4
2.2 Light Organ Lure........................................................................................................ 10
2.3 Intraspecific Communication ..................................................................................... 12
2.4 Camouflage ................................................................................................................. 12
2.5 Searchlight .................................................................................................................. 13
2.6 Functionless Bioluminescence.................................................................................... 14
3 Terrestrial Taxa ................................................................................................................... 14
3.1 On Land ...................................................................................................................... 14
1 Introduction
2 Function of Bioluminescence
2.1 Self-defense
Many different biological and ecological functions have been proposed for bio-
luminescence [1, 2], but the most parsimonious explanation is that it is used as an
antipredator defensive device. Here we categorize the various defensive reactions
by luminous organisms aimed at predators into three types: startling (or deterring),
misdirection (or distraction/obstruction of sight), and aposematism (warning
display).
Eco-Evo Bioluminescence on Land and in the Sea 5
Fig. 1 The diversity of bioluminescent organisms as proportions of the named 746 genera. The
fraction indicates the number of terrestrial genera relative to the total number of genera. Numbers
are based on Herring [1] and the supplemental material in Haddock et al. [2]. The genera not
certain to be luminous in Herring [1], and ‘‘doubtfully’’ luminous genera of sponge and bryzoan
[2] are excluded. Numbers of genera in some groups are modified according to recent
publications: Bacteria [139], Lampyridae [173], Phengodidae [8], Rhagophthalmidae [174],
Gastropoda [109], Oligochaeta [102], Homalisidae (Omalisidae) [81], and Fungi [97]. Nematode
Heterorhabditis is excluded because symbiotic luminous bacteria Photorhabdus does not emit
visible light in the nematode [100]. For compatibility with the list by Herring [1], the
classification of Division and Phylum is followed in the systematics by Parker [95]
Sudden flashes in dark surroundings have been shown to startle, deter, and stun
or temporarily blind predators regardless of the prey’s palatability. For example,
the mesopelagic squid, Taningia danae, uses its arm-tip photophores to flash in
bursts while attacking bait rigs [3]. Moreover, in the firefly squid Watasenia
scintillans (Fig. 2), the nervous-system controlled arm-tip photophores are thought
to flash in bursts when attacked by predators [4, 5]. This seems to be similar to
cases in which the eye-spots on the surfaces of the hind wings of some Lepidoptera
startle predators by their sudden exposure [6], an example of Batesian mimicry in
which reflective owl eyes are the model. However, startling bioluminescence is
probably not Batesian mimicry, as it does not imitate any predator models. The
exceptions include some phengodid beetle larviform female adults, and the adults
of click beetles. The female phengodids Diplocladon and Rhagophthalmus curl
their bodies around their eggs to protect the brood, and when disturbed emit light
from small circular photophores lining the body (Fig. 3).
6 Y. Oba and D. T. Schultz
Fig. 2 Firefly squid Watasenia scintillans collected in Toyama Bay, Japan. This picture shows
the interspersed photophores on the ventral side of the mantle that face down as the creature
swims. The larger light organ on the tip of arms is assumed to be used for stunning predators.
Photo courtesy of So Yamashita
Fig. 3 Luminescence of the phengodid beetle Rhagophthalmus ohbai female adult at Ishigaki
Island, Japan. Attracting a male (left) and after copulation (right)
Ohba [7] hypothesized that the light emission may function to startle predators
by mimicking the shining eyes of nocturnal birds. The adults of the luminous click
beetles (e.g., Jamaican Pyrophorus species) possess a pair of oval photophores at
the outward-angled posterior margins of the prothorax [8], and the luminescence
looks like the shining eyes of nocturnal animals found in forests at night (VB
Meyer-Rochow, personal communication). Thus, the luminescence of phengodid
and click beetles may be an example of a startling function evolved in the presence
of a sympatric predator model.
Eco-Evo Bioluminescence on Land and in the Sea 7
Fig. 6 Luminous fungus Mycena chlorophos fruiting body damaged by ants at nighttime in situ
on Hachijo Island, Japan. Photo courtesy of Masashi Naito
Fig. 7 Luminous fireworm Odontosyllis sp. after autotomy by stimulation. Detached tail tip
glows continuously. Specimens were collected in Toyama Bay, Japan. Photo courtesy of Masashi
Naito
Deep-sea anglerfish, such as Himantolophus, are known to attract prey with a light
organ created by cultivating symbiotic luminous bacteria in the tips of their escas,
that is, the lure-like structure created from the modified first spine of the dorsal fin.
However, anglerfish have not been observed in situ luring prey using lumines-
cence. Using bioluminescence to attract prey has also been suggested for other
marine animals, such as stomiid dragonfish, Chiroteuthis squid, Stauroteuthis
octopus, and Siphonophores [2].
There are some hypotheses as to why prey are attracted to luminescence in the
ocean. One plausible explanation is that luminescent particles in the sea visually
indicate food. For example, deep-sea marine snow contains organic detritus and
fecal pellets that host glowing bacteria. Small zooplanktons and fry detect the
luminescence and consume the luminous material [39], and consequently, zoo-
plankton are attracted to the luminescence. Zarubin et al. [39] suggests that the
luminescence of nonsymbiotic bacteria enables them to find a host and to prop-
agate in the nutrient-rich intestines of zooplankton and fish. Previous studies
corroborate the high level of nonsymbiotic luminous bacteria in fecal pellets [40,
41].
The caves and riverbanks of Australia and New Zealand are home to larvae of
fungus gnats, Arachnocampa spp., that attract small phototactic insects with
luminescent posterior light organs into nonluminescent snares of vertical threads
(termed ‘‘fishing-lines’’) coated with mucous [42–44] (Fig. 8). A similar behavior
Eco-Evo Bioluminescence on Land and in the Sea 11
was also reported from the North American fungus gnat Orfelia fultoni [45, 46].
Interestingly the larvae of fungus gnats of the genus Keroplatus (Fig. 9), are also
luminescent, despite that these species are likely to be spore-feeders [47]. The
luminescence function of Keroplatus is unclear [9].
Most fireflies do not feed in adult stages. By contrast, it is very unique and
exceptional that female adults of the North American fireflies Photuris and Bi-
cellonycha mimic flash patterns of other fireflies, for example, Photinus and
Pyractomena females, thereby attracting the males of the latter as prey [48].
Photuris cannot produce toxins by themselves, so they obtain the toxins, rather
than nutrition, from prey fireflies and use them for aposematic display [49–51].
Larvae of the luminous click beetle Pyrearinus termitilluminans reside in termite
mounds in Central America, and emit light from their prothorax to attract alate
ants and termites as prey [52].
One of the roles of luminescence in mushrooms may be the attraction of spore-
dispersing organisms. This hypothesis has been widely accepted for many years,
but has never clearly been demonstrated [34].
12 Y. Oba and D. T. Schultz
Luminescence intended for prey sometimes attracts predators instead. The cave
harvestmen Megalopsalis and Hendea possess prominent eyes to detect the
luminescence of larval Arachnocampa luminosa, which are their major prey [53].
It is hypothesized that some large-sized marine predators such as sperm whales
and leatherback turtles may use bioluminescent cues in their search for their food
([2], and references therein).
On land, it is well known that some adult fireflies use luminescence signals for
sexual communication between males and females [51, 54, 55]. Larviform females
of R. ohbai display their dorsal photophore toward males with continuous lumi-
nescence (Fig. 3), suggesting the function of luminescence is sexual communi-
cation [55]. The luminescence of the females of other phengodid beetles,
Dioptoma and Diplocladon, also functions to attract their respective males [54,
56]. The function of luminescence in luminous click beetles has also been sug-
gested as a means of sexual communication between males and females [4, 57].
Indeed, the male adults are sometimes attracted to artificial lights, including cig-
arettes [58]. The adults of the New Zealand fungus gnat Arachnocampa luminosa
may use luminescence for sexual communication [42, 59, 60]; this has been
challenged by Broadley [61], who provides evidence in support of pheromonal
attraction.
In the ocean, bioluminescence in some ostracods, including the genus Vargula
and the polychaete genus Odontosyllis has been suspected to possess a role in
sexual communication based on observations of their behavior in the wild [4, 10,
38]. Luminescence of certain shallow water fish, such as leiognathid ponyfish and
the flashlight fish Photoblepharon palpebratus, may function as a means of
intraspecific communication (for mating, dominance display, and/or schooling)
[62–64]. Based on some sexual dimorphism in photophores (light organs) it has
been suggested that various midwater organisms use luminescence for sexual
communication [65, 66].
2.4 Camouflage
Fig. 10 Midwater
hatchetfish Sternoptyx
pseudobscura, with ventral
photophores for counter-
illumination and large eyes
for finding the silhouettes of
prey by looking up. Photo
courtesy of Laboratory of
Marine Biology, Kochi
University
turtles, whales, and sharks. On the other hand, smaller animals are at risk of attack
for many predators look up to find prey (Fig. 10).
Dahlgren [67] first suggested that ventral light organs in luminous squid are
used to negate their silhouette by shining at ambient brightness levels. This
counter-illumination hypothesis has also been used to explain luminescence in
various luminous fish and shrimp; some of their downward-pointing light organs
were shown to dynamically match background light intensity in laboratory studies
[68, 69].
Counter-illumination is common in marine mesopelagic dwellers [70] but not
supported in terrestrial and freshwater species. The myriad shelters and hiding
spots on land as well as in the shallow or cloudy waters of rivers, ponds, and lakes
may in the course of evolution have led to alternatives to bioluminescence as a
survival strategy.
2.5 Searchlight
Visible light from some organisms may not have any significance in itself. Bio-
luminescence is considered to be an energetically low-cost mechanism [75, 76],
and thus can potentially evolve as a metabolic byproduct unless the light emission
is disadvantageous. For example, it has been proposed that fungal luminescence is
a byproduct of some unknown metabolic process [34, 77]. Luminescence of
organisms emitting very weak light, such as the Japanese fungus gnat Keroplatus
nipponicus, or glowing underground, such as the annelid Microscolex phospho-
reus, may also be functionless.
3 Terrestrial Taxa
3.1 On Land
vertical threads using luminescence, it seems strange that no luminous spider has
been reported to date.
In the subphylum Myriapoda, some millipedes (Diplopoda) and centipedes
(Chilopoda) emit light, with 24 known species altogether [25]. In the subphylum
Hexapoda, luminous species are only found in elateroid beetles (Coleoptera),
fungus gnats (Diptera), and springtails (Collembola) [9, 81].
Coleoptera are the largest order in the class Insecta, containing about 350,000
species in over 160 families worldwide [22]. Luminescent species of beetles are
found in three elateroid families: Lampyridae, Phengodidae, and Elateridae [81,
82]. All species of the Lampyridae and Phengodidae (roughly 2,000 and 200
species worldwide, respectively) are luminous, and of the approximately 10,000
elaterid species worldwide, approximately 200 are luminous [83].
The Diptera (true flies) are the fourth largest order in the Insecta, containing
150 families and 150,000 species [84]. Despite their huge diversity, only a small
number of luminous species (*15 species) are known from a single family, the
Keroplatidae [9]. The bioluminescence mechanism of Arachnocampa fungus gnats
is considered to be different from that of Orfelia [85], but the details are still
unknown.
In Collembola, luminescent species have been recognized in the families
Neanuridae and Onychiuridae [4, 56], but the observation records are limited. The
author (YO) observed the luminescence of Lobella sp. (Neanuridae), in which the
specimens emitted a continuous weak light from the body after stimulation with
forceps. The function of luminescence in Lobella sp. is uncertain but it seems too
faint to deter predators or to use for intraspecific communication [9].
Recently luminescence was claimed to occur in some South American cock-
roaches (Blattodea) [86, 87], but the veracity of these reports is highly dubious [88].
It is interesting that Lepidoptera, the second largest order in Insecta [22]
apparently possess no luminous species. Most adult moths are nocturnal and some
are poisonous; thus bioluminescence would seem a suitable tool for communica-
tion by moths. However, lepidopteran insects do not seem to have evolved any
bioluminescence and instead widely use pheromones for their nocturnal commu-
nications. It seems worth noting that some nonluminous fireflies also use phero-
mones for intraspecific communication, but in contrast to the moths, the beetles are
diurnal. In Hemiptera, the fifth largest insect order [22], the elongated head of the
lantern fly Fulgora laternaria in South America had long been believed to be
luminescent, but this has now become very doubtful [89].
Luminous species of the three elateroid beetle families use the same luciferin
molecule in their luminescence mechanism, (4S)-4,5-dihydro-2-(6-hydroxy-2-
benzothiazolyl)-4-thiazolecarboxylic acid, and a homologous luciferase sharing
over 48 % amino acid identity [90]. Phylogenetic relationships among these fam-
ilies have been studied in the context of the evolution of bioluminescence [18, 91],
but are still not fully resolved. Recent molecular analysis based on the mitochon-
drial genome revealed that Lampyridae and Phengodidae are sister groups in the
cantharoid beetles [92], suggesting a single origin of their luminescence. Although
Elateridae are also closely related to the Lampyridae–Phengodidae clade [92],
16 Y. Oba and D. T. Schultz
phylogenetic analyses of Elateridae showed that luminous click beetles are deeply
nested within the nonluminous species, thus bioluminescence may have arisen
independently in Elateridae [93, 94]. These results indicate that bioluminescence in
beetles may have evolved once or twice [81].
Bioluminescence mechanisms of the land snail Quantula, millipedes, centi-
pedes, fungus gnats, and springtails are still unknown in detail ([9], and references
therein), but probably are independent from each other. Therefore, this author
(YO) suspects that bioluminescence has evolved at least seven times among ter-
restrial organisms, including the case of luminous fungi described hereafter.
Luminous fungi are found only in the phylum Basidiomycota (Division
Eumycota, sensu Parker [95]), that is, the taxon of fungi that produces large,
apparent fruiting bodies referred to as ‘‘mushrooms’’ (approximately 32,000 spe-
cies [96]). About 70 species of luminescent fungi have been recognized world-
wide, and they belong to three different subclades: Omphalotus, Armillaria, and
the mycenoid fungi [97]. Although all fungal luminescence is intracellular, some
species glow only in the mycelia (e.g., Armillaria species), whereas others lumi-
nesce in both mycelia and fruiting bodies (e.g., O. japonicus and M. chlorophos).
Even though the bioluminescence mechanism of the luminous fungi is not fully
understood [98], the reaction mechanism is known to be identical in all species
[99]. The function of bioluminescence in fungi has long been questioned, and
remains unconfirmed experimentally.
Terrestrial luminescent bacteria of the genus Photorhabdus adhere to the
intestine of a Heterorhabditis nematode worm: the nematode burrows into its
insect host and regurgitates Photorhabdus into the hemocoel; the bacteria grow in
the insect body and finally kill the insect. The Nematode feeds exhaustively on the
bacteria or products of bacterial degradation of the hemolymph then leaves the
cadaver in search of a new host insect. The luminescence of Photorhabdus is
observed only when it grows in the insect cadaver. The ecological functions of the
cadaver luminescence has been uncertain, but one suggestion was that it may
represent an aposematic warning to nocturnal scavenging animals or act as a lure
to attract further insect prey [100].
Some lampyrid beetles are well known for luminescing during flight, and are
rightly commonly called fireflies. However, not all luminous beetles are capable of
flight. Some phengodid (e.g., Rhagophthalmus) and lampyrid beetles (e.g.,
Lampyris) have luminous but flightless wingless females (Fig. 3) and are com-
monly called glow-worms, whereas the males are capable of flight (winged) but
are nonluminous or only very dimly luminescent [20, 55]. Male and female adults
of some lampyrid genera are able to fly but are nonluminous, such as Lucidota,
Pyropyga, and Pristolycus [101]. The manipulation of luminescence in some
winged fireflies, such as in Luciola and Photinus, is very sophisticated: they use
Eco-Evo Bioluminescence on Land and in the Sea 17
species-specific flash signals to recognize conspecific males and females [51, 82,
101]. All luminous click beetles are likely capable of flight as adults, and it has
been suggested that these males and females use the light for sexual communi-
cation while flying [57, 101].
Compared with the rich diversity of luminous organisms in the sea, luminous
freshwater dwellers are quite rare. Fish and crustaceans make up a large per-
centage of the list of luminous organisms [1, 105], but there are apparently no
luminescent freshwater fish and crustacean species.
The limpet Latia neritoides (Latiidae, Mollusk) found in streams of New
Zealand’s North Island is the world’s only luminous freshwater gastropod species.
18 Y. Oba and D. T. Schultz
crayfish, or dragonfly larvae, and suggested ‘‘the predators may chase after
luminescent droplets washed away from Latia instead of attacking Latia itself’’.
Most of the lampyrid species are terrestrial for all phases of their lifecycle, but
only a few Luciola species (*10 species) are known to be aquatic during the
larval stage [107]. They adapt well to the aquatic environment where they locate
and overpower freshwater snails in order to feed on them. Most, but not all, of the
aquatic species’ larvae respire in water using tracheal gills [16, 107]. These aquatic
larvae project white glands from abdominal segments that emit a fetid smell,
suggesting that the luminescence of some aquatic larvae is also an aposematic
display to predators, such as freshwater goby and dragonfly larvae [14].
4 Marine Taxa
The intertidal area between the sea and land, called the littoral zone, is regularly
exposed to variable conditions: rain causes low salinity, and sunlight causes heat,
desiccation, strong UV/visible light, and high salinity. Only a few shellfish and an
earthworm are reported as luminous in this environmentally challenging zone.
Some tiny luminous gastropod snails, Angiola and Hinea (Planaxidae), can be
found under rocks in tide pools at low tide and on stony beaches [108–111]. The
luminous bivalve Pholas dactylus inhabits the shallow sublittoral zone of soft-rock
coasts, typically gneiss [4]. It is of interest that the luminous bivalves and
20 Y. Oba and D. T. Schultz
gastropod snails are found only in the harsh intertidal zone, given that there are
many thousands of related species that inhabit almost every depth of the ocean.
One possible exception is the eulimid gastropod snails Melanella luminosa, which
was found as a parasite on the scarlet sea cucumber at a depth of 10–238 m;
Marshall [112] has described its yellow glow.
In the snail Angiola zonata (syn. Angiola longispira) (Fig. 14), a patch on the
mantle (or hypobranchial gland, [109]) emits a weak blue-green light when the
animal is stimulated mechanically or exposed to high salinity ([108], S Yamashita,
personal communication). Houbrick [109] reported that all species in Angiola
appear to be nocturnal and suggested that the light in ‘‘Angiola species may have a
startling effect on predators’’. Deheyn and Wilson [111] suggested that an apo-
sematic function ‘‘is less probable considering the gregarious and cryptic behav-
iour’’ of planaxid snails. The author (YO) observed in aquaria that fry of the
intertidal teleost Kuhlia mugil consumed the soft bodies of A. zonata with no
apparent ill effect, suggesting the snail’s luminescence is not for aposematic dis-
play. In Pholas, luminescent mucus is discharged from the siphon when disturbed,
and as Harvey suggested, ‘‘the best guess is that the light masses scare away
predacious animals’’ [4], but the actual function remains uncertain [37].
The luminous earthworm Pontodrilus litoralis (syn. P. matsushimensis, P.
bermudensis) inhabits the littoral zone of sandy beaches. The function of biolu-
minescence in P. litoralis remains unclear, likewise in other luminous earthworms.
In Japan, this species is sometimes used as bait for fishing [113]. Furthermore, a
crab and goby immediately fed on living P. litoralis when introduced into aquaria,
and no apparent ill effects in the predators were observed after consuming the
luminescent earthworm, suggesting it is neither distasteful nor toxic for potential
predators. The author (YO) has observed carnivorous insects such as earwigs
(Dermaptera) and staphylinid beetles (Staphylinidae) occurring sympatrically with
P. litoralis. These observations suggest that the function of P. litoralis lumines-
cence might be to startle, but not as an aposematic display. On the other hand, as
luminous fluid is discharged only when P. litoralis specimens are severely stim-
ulated, such as being cut, squeezed, and/or beaten [9, 114], the startling hypothesis
will need to be verified.
Eco-Evo Bioluminescence on Land and in the Sea 21
In the neritic zone, the number of luminescent species is relatively low, a reported
1–2 % of all coastal species [115]. However, each luminous species often occurs
in high density in its environment. This situation is in contrast to those in oceanic
waters where species diversity is very high, but population density of individual
species is usually low (see Sect. 4.3). Luminous species usually make up only a
small proportion of a phylum’s total number of species, but there are two
exceptions: the Cnidaria (jellyfish) and the Ctenophora (comb jelly). A large
proportion of the former phylum and nearly all of the latter are luminous [4], and
these two groups are common members of the coastal planktonic dwellers. As for
the biological function of luminescence in coastal dwellers, Morin [115] suggested
that these species primarily use fast flashes of luminescence to deter prey or
continuously glowing decoys to distract predators, giving the prey an opportunity
to escape.
Dinoflagellates (Division Chromophycota, sensu Parker [95]) are important
primary producers in coastal waters. Some of the free-living species are luminous,
and most of these are autotrophic and therefore live in shallow waters where they
can photosynthesize. The luminous dinoflagellate Noctiluca scintillans is an
obligate heterotroph, but it also lives in shallow water to feed on autotrophic
diatoms, other dinoflagellates, and even small zooplankton [37]. For the enzymatic
bioluminescence reaction, luminous dinoflagellates use open-chain tetrapyrrole
luciferin [116], which is structurally related to chlorophyll. Interestingly, luminous
krill (Euphausiidae) also use the tetrapyrrole luciferin, which is structurally very
similar to the luciferin of dinoflagellates [117, 118]. Accordingly, it is expected
that krill consume luminous dinoflagellates and chemically modify the dinofla-
gellate luciferin as a precursor to their own luciferin [119]. To date, other luminous
taxa that use dinoflagellate luciferin or its derivatives as a luminous substrate have
not been reported.
In ostracods, luminous species in the genera Vargula and Cypridina are coastal,
whereas species in the genus Conchoecia are midwater dwellers [120]. The two
former genera use cypridinid luciferin (also called Cypridina luciferin) as a bio-
luminescent substrate [98, 121], whereas Conchoecia use coelenterazine [98, 122].
These findings are consistent with the fact that no midwater fish are known that use
cypridinid luciferin as a bioluminescent substrate (see below).
Coastal fish with nonbacterial luminescence rely on cypridinid luciferin in
bioluminescence reactions [123, 124]. These types of luminous fish are found in
three distantly related families of the orders Perciformes and Batrachoidiformes
[123–128]: Parapriacanthus ransonneti (syn. P. beryciformes [129]) and Par-
apriacanthus dispar (Pempheridae); Pempheris ypsilychnus (Pempheridae [130]);
22 Y. Oba and D. T. Schultz
Apogon ellioti (syn. Jaydia ellioti) and Apogon poecilopterus (syn. Jaydia poe-
cilopterus) (Apogonidae); midshipman fish Porichthys spp. (P. notatus, myriaster,
and porosissimus; Batrachoididae); and presumably some Archamia and Rhabd-
amia species (Apogonidae).
It has been suggested that these coastal fish obtain cypridinid luciferin by
consuming Cypridina and Vargula ostracods [37]. In fact, ostracod specimens
have been discovered from the stomach of P. ransonneti ([131], Oba and Hiruta,
unpublished), and a nonluminous form of P. notatus recovered its luminescent
ability following injection of cypridinid luciferin [132]. Recently, the author (YO)
and colleagues demonstrated that cypridinid luciferin is biosynthesized from three
amino acids (Ile, Arg, Trp) in Vargula hilgendorfii (Fig. 4) and Cypridina noc-
tiluca [133–136]. These findings indicate that cypridinid luciferin produced by
coastal ostracods partly contributes to the species’ richness of luminous fish in the
coastal environment (Fig. 15). Indeed, cypridinid luciferin chemiluminesces
strongly in certain solvents, such as diglyme, dimethyl sulfoxide, and detergent-
water solution, without an enzymatic catalyst [137]. This high reactivity suggests
that cypridinid luciferin is potentially a good bioluminescent substrate by nature.
Therefore, it may not be difficult to evolve luciferase from an extant protein, if
organisms are able to obtain cypridinid luciferin by diet.
Eco-Evo Bioluminescence on Land and in the Sea 23
Previous research suggests that nearly all of the luminescent species known to
occur in the midwater of oceans. Beebe [138] estimated that based on a study in
Bermuda that luminous mesopelagic fish collected below a depth of 600 m rep-
resent 81 % of the families, 66 % of the species, and 97 % of all the individuals.
Calanoid copepods, krill, and Cyclothone fish represent a large part of total macro-
and microzooplanktonic marine biomass, and these taxa contain particularly many
luminous species [4].
Haddock et al. [2] listed hypotheses of why most luminescent taxa are ocean
dwellers: they suggested that the sea is favorable for the evolution of biolumi-
nescence because it is (1) a stable environment, (2) optically clear relative to other
aqueous habitats, (3) continuously dark or very dim, and (4) highly diversified
taxonomically with complex interspecific relationships. We suggest that there are
additional factors that favor the evolution or acquisition of bioluminescent capa-
bilities in marine organisms: namely ‘‘symbiotic luminescence’’ and ‘‘dietary
supply of coelenterazine’’.
4.3.2 Coelenterazine
Based on a recent study in the Bahamas, the benthic zone of the deep sea
(500–1,000 m depth) is not as species-rich in luminous organisms as the midwater
zone: less than 20 % of the collected species emitted light [160]. Johnsen et al.
[160] explain that visibility in benthic water is obstructed by particulate matter
stirred by a bottom current, which is in contrast to the optically clear midwater that
facilitates the evolution of bioluminescence [2].
In the deep-sea benthos, luminous species are found mostly in bamboo corals,
sea anemones, sea pens, sea cucumbers, and brittle stars [160]. Because it is
extremely difficult to observe natural behavior of the organisms, the functions of
their luminescence are still unclear. There are many bottom dwellers in crabs,
hermit crabs, lobsters, and isopods, but no luminous species have been reported in
these groups. An unidentified luminous sea anemone was found on the shell
inhabited by a hermit crab [160]. It is known that some sea anemones and hermit
crabs engage in a mutualistic symbiosis [161]. If the deep-sea hermit crab posi-
tively selects luminous sea anemones as a symbiont, the hermit crab should be
Eco-Evo Bioluminescence on Land and in the Sea 27
5 Conclusion
physical stimulation, or the light may be for sexual communication given that
morphological variations of photophores between sexes are rather common. It is,
however, best to be wary of cursory postulations: for example, stimulation-induced
lights may be too weak to startle predators and photophore sexual dimorphism may
be involved in behaviors other than mating. Meanwhile, some luminous organisms
possess highly sophisticated light organs (equipped with lenses, color filters,
reflectors, diffusers), and are neuronally controlled [168], suggesting that the light
must have some biological significance, albeit poorly understood. Moreover, some
unique hypotheses such as ‘‘predatory counter-illumination’’ for luminous sharks
[169], and ‘‘luminescent burglar alarms’’ [115, 164] for luminous dinoflagellates
and luminous fungi ([170] and [34], respectively) may illuminate wealthiness of
bioluminescence when tested experimentally in future.
6 Perspectives
This chapter has presented an ecological and evolutionary overview of the many
divergent bioluminescent taxa found in the world. One important observation has
been the great diversity of bioluminescent substrates and enzymes that has arisen
during the evolution of the varied taxa. Moreover, the paths of convergent evo-
lution have yielded a wide variety of ecological behaviors that utilize
bioluminescence.
Although the luciferin structures of many organisms were resolved between the
1960s and 1980s, many luciferase or photoprotein genes remain unknown, such as
those of euphausiid krill Euphausia pacifica, the firefly squid W. scintillans
(Fig. 2), the midwater lanternfish of the family Myctophidae, the shallow water
fish Parapriacanthus and Porichthys, the earthworm Diplocardia longa, and the
freshwater limpet L. neritoides. Several taxa still have eluded identification of both
their luciferin and their luciferase, including the millipedes Paraspirobolus and
Motyxia, the polychaete Odontosyllis, and the brittle star Ophiopsila (summarized
in [98]). In the case of luminous fungi, enzymatic involvement in light production
is still unresolved [98, 171].
To facilitate future research of bioluminescent organisms, my group is pro-
ceeding with a DNA barcoding initiative to create a genetic reference library for
bioluminescent organisms, per Hebert et al. [172]. The database will provide a
resource for other scientists in the identification of luminous organisms with
unresolved bioluminescent systems. Given the many applications of biolumines-
cent systems using only a few well-characterized protein families, elucidating
currently unknown proteins, genes, and substrates will expand the toolkit of
molecular science and bioengineering. As there are likewise many species for
which the biological and ecological function of bioluminescence is poorly
understood or disputed in the literature, this same principle will apply to ecologists
who seek to study bioluminescent organisms.
Eco-Evo Bioluminescence on Land and in the Sea 29
References
20. Branham MA, Wenzel JW (2003) The origin of photic behavior and the evolution of sexual
communication in fireflies (Coleoptera: Lampyridae). Cladistics 19:1–22
21. De Cock R, Matthysen E (1999) Aposematism and bioluminescence: experimental evidence
from glow-worm larvae (Coleoptera: Lampyridae). Evol Ecol 13:619–639
22. Grimaldi D, Engel MS (2005) Evolution of the insects. Cambridge University Press,
Cambridge
23. Sivinski J (1981) The nature and possible functions of luminescence in Coleoptera larvae.
Coleopterists Bull 35:167–179
24. Raj JS (1957) An undescribed luminous beetle larva from South India. J Bombay Natl Hist
Soc 54:788–789
25. Rosenberg J, Meyer-Rochow VB (2009) Luminescent myriapoda: a brief review. In: Meyer-
Rochow VB (ed) Bioluminescence in focus: a collection of illuminating essays. Research
Signpost, Kerala, pp 139–146
26. Marek P, Papaj D, Yeager J, Molina S, Moore W (2011) Bioluminescent aposematism in
millipedes. Cur Biol 21:R680–R681
27. Davenport D, Wootton DM, Cushing JE (1952) The biology of the Sierra luminous
millipede, Luminodesmus sequoiae, Loomis and Davenport. Biol Bull 102:100–110
28. Hudson BJ, Parsons GA (1997) Giant millipede ‘burns’ and the eye. Trans Roy Soc Trop
Med Hygiene 91:183–185
29. Houdemer ME (1926) Mote sur un Myriapode vésicant du Tonkin, Ostostigmus aculeatus
Haase. Bull Mus Hist Nat Paris 32:213–214 (in French)
30. Futuyma DJ (2005) Evolution. Sinauer, Massachusetts
31. Ohba N, Meyer-Rochow VB (2012) Insect species co-existing with the Papua New Guinea
firefly Pteroptyx effulgens share aspects of appearance and behaviour. Lampyrid 2:127–137
32. Crowson RA (1981) The biology of the Coleoptera. Academic Press, New York
33. Lev-Yadun S, Halpern M (2007) Ergot (Claviceps purpurea)—An aposematic fungus.
Symbiosis 43:105–108
34. Sivinski J (1981) Arthropods attracted to luminous fungi. Psyche 88:383–390
35. Mallefet J (2009) Echinoderm bioluminescence: where, how and why do so many
ophiuroids glow? In: Meyer-Rochow VB (ed) Bioluminescence in focus: a collection of
illuminating essays. Research Signpost, Kerala, pp 67–83
36. Grober MS (1988) Brittle-star bioluminescence functions as an aposematic signal to deter
crustacean predators. Anim Behav 36:493–501
37. Wilson T, Hastings JW (2013) Bioluminescence: living lights, lights for living. Harvard
University Press, Massachusetts
38. Abe K (1994) The light of marine fireflies. Chikuma Shobo, Tokyo (in Japanese)
39. Zarubin M, Belkin S, Ionescu M, Genin A (2012) Bacterial bioluminescence as a lure for
marine zooplankton and fish. Proc Natl Acad Sci USA 109:853–857
40. Andrews CC, Karl DM, Small LF, Fowler SW (1984) Metabolic activity and
bioluminescence of oceanic faecal pellets and sediment trap particles. Nature 307:539–541
41. Ruby EG, Morin JG (1979) Luminous enteric bacteria of marine fishes: a study of their
distribution, densities, and dispersion. Appl Environ Microbiol 38:406–411
42. Richards AM (1960) Observations on the New Zealand glow-worm Arachnocampa
luminosa (Skuse) 1890. Tran R Soc New Zealand 88:559–574
43. Broadley A, Stringer IAN (2009) Larval behaviour of the New Zealand glowworm,
Arachnocampa luminosa (Diptera: Keroplatidae), in bush and caves. In: Meyer-Rochow VB
(ed) Bioluminescence in focus: a collection of illuminating essays. Research Signpost,
Kerala, pp 325–355
44. Willis RE, White CR, Merritt DJ (2011) Using light as a lure is an efficient predatory
strategy in Arachnocampa flava, an Australian glowworm. J Comp Physiol B 181:477–486
45. Fulton BB (1941) A luminous fly larva with spider traits (Diptera, Mycetophilidae). Ann
Entomol Soc Am 34:289–302
46. Sivinski J (1982) Prey attraction by luminous larvae of the fungus gnat Orfelia fultoni. Ecol
Entomol 7:443–446
Eco-Evo Bioluminescence on Land and in the Sea 31
47. Matile L (1997) Phylogeny and evolution of the larval diet in the Sciaroidea (Diptera,
Bibionomorpha) since the Mesozoic. Mém Mus Natn Hist Nat 173:273–303
48. Lloyd JE (1975) Aggressive mimicry in Photuris fireflies: signal repertories by femmes
fatales. Science 187:452–453
49. Eisner T, Goetz MA, Hill DE, Smedley SR, Meinwald J (1997) Firefly ‘‘femmes fatales’’
acquire defensive steroids (lucibufagins) from their firefly prey. Proc Natl Acad Sci USA
94:9723–9728
50. Eisner T, Wiemer DF, Haynes LW, Meinwald J (1978) Lucibufagins: defensive steroids
from the fireflies Photinus ignitus and P. marginellus (Coleoptera: Lampyridae). Proc Natl
Acad Sci USA 75:905–908
51. Lewis SM, Cratsley CK (2008) Flash signal evolution, mate choice, and predation in
fireflies. Annu Rev Entomol 53:293–321
52. Redford KH (1982) Prey attraction as a possible function of bioluminescence in the larvae
of Pyrearinus termitilluminans (Coleoptera: Elateridae). Revta bras Zool S Paulo 1:31–34
53. Meyer-Rochow VB, Liddle AR (1988) Structure and function of the eyes of two species of
opilionid from New Zealand glow-worm caves (Megalopsalis tumida: Palpatores, and
Hendea myersi cavernicola: Laniatores). Proc R Soc B 233:293–319
54. Lloyd JE (1983) Bioluminescence and communication in insects. Ann Rev Entomol
28:131–160
55. Ohba N (2004) Flash communication systems of Japanese fireflies. Integ Comp Biol
44:225–233
56. Haneda Y (1985) Luminous organisms. Kouseisha-kouseikaku, Tokyo (in Japanese)
57. Stolz U, Velez S, Wood KV, Wood M, Feder JL (2003) Darwinian natural selection for
orange bioluminescent color in a Jamaican click beetle. Proc Natl Acad Sci USA
100:14955–14959
58. Hoffmann KH (1984) Environmental aspects of insect bioluminescence. In: Hoffmann KH
(ed) Environmental physiology and biochemistry of insects. Springer, Berlin, pp 225–245
59. Meyer-Rochow VB, Eguchi E (1984) Thoughts on the possible function and origin of
bioluminescence in the New Zealand glowworm Arachnocampa luminosa (Diptera:
Keroplatidae), based on electrophysiological recordings of spectral responses from the
eyes of male adults. New Zealand Entomol 8:111–119
60. Meyer-Rochow VB (1990) The New Zealand glowworm. Waitomo Caves Mus, Waitomo
Caves
61. Broadley RA (2012) Notes on pupal behaviour, eclosion, mate attraction, copulation and
predation of the New Zealand glowworm Arachnocampa luminosa (Skuse) (Diptera:
Keroplatidae), at Waitomo. N Zld Ent 35:1–9
62. Morin JG, Harrington A, Nealson K, Krieger N, Baldwin TO, Hastings JW (1975) Light for
all reasons: Versatility in the behavioral repertoire of the flashlight fish. Science 190:74–76
63. Meyer-Rochow VB (1976) Womit und warum Tiere leuchten. Selecta 10:972–974 (in
German)
64. Chakrabarty P, Davis MP, Smith WL, Berquist R, Gledhill KM, Frank LR, Sparks JS (2011)
Evolution of the light organ system in ponyfishes (Teleostei: Leiognathidae). J Morphol
272:704–721
65. Young RE (1983) Oceanic bioluminescence: an overview of general functions. Bull Mar Sci
33:829–845
66. Herring PJ (2007) Sex with the lights on? A review of bioluminescent sexual dimorphism in
the sea. J Mar Biol Ass UK 87:829–842
67. Dahlgren U (1916) Production of light by animals. J Franklin Inst 181:525–556
68. Young RE, Roper CFE (1977) Intensity regulation of bioluminescence during
countershading in living midwater animals. Fish Bull 75:239–252
69. Case JF, Warner J, Barnes AT, Lowenstine M (1977) Bioluminescence of lantern fish
(Myctophidae) in response to changes in light intensity. Nature 265:179–181
70. Widder EA (2010) Bioluminescence in the ocean: origins of biological, chemical, and
ecological diversity. Science 328:704–708
32 Y. Oba and D. T. Schultz
71. Hunt DM, Dulai KS, Partridge JC, Cottrill P, Bowmaker JK (2001) The molecular basis for
spectral tuning of rod visual pigments in deep-sea fish. J Exp Biol 204:3333–3344
72. Widder EA, Latz MI, Herring PJ, Case JF (1984) Far red bioluminescence from two deep-
sea fishes. Science 225:512–514
73. Douglas RH, Partridge JC, Dulai KS, Hunt DM, Mullineaux CW, Hynninen PH (1999)
Enhanced retinal longwave sensitivity using a chlorophyll-derived photosensitiser in
Malacosteus niger, a deep-sea dragon fish with far red bioluminescence. Vis Res
39:2817–2832
74. Meyer-Rochow VB, Baburina V, Smirnov S (1982) Histological observations on the eyes of
the two luminescent fishes Photoblepharon palpebratus (Boddaert) and Anomalops
katoptron (Blkr.). Zool Anz (Jena) 209:65–72
75. O’Kane DJ, Lingle WL, Porter D, Wampler JE (1990) Spectral analysis of bioluminescence
of Panellus stypticus. Mycologia 82:607–616
76. Woods WA Jr, Hendrickson H, Mason J, Lewis SM (2007) Energy and predation costs of
firefly courtship signals. Am Nat 170:702–708
77. Herring PJ (1994) Luminous fungi. Mycologist 8:181–183
78. Haneda Y (1963) Further studies on a luminous land snail, Quantula striata, in Malaya. Sci
Rept Yokosuka City Mus 8:1–9
79. Counsilman JJ, Ong PP (1988) Responses of the luminescent land snail Dyakia (Quantula)
striata to natural and artificial lights. J Ethol 6:1–8
80. Noll F (1888) Über das Leuchten der Schistostega osmundacea Schimp. Arbeiten Bot Inst
Würzburg 3:477–488 (in Germany)
81. Oba Y (2009) On the origin of beetle luminescence. In: Meyer-Rochow VB (ed)
Bioluminescence in focus: a collection of illuminating essays. Research Signpost, Kerala,
pp 277–290
82. Lloyd JE (1978) Insect bioluminescence. In: Herring PJ (ed) Bioluminescence in action.
Academic Press, New York
83. Costa C, Lawrence JF, Rosa SP (2010) Elateridae Leach, 1815. In: Leschen RAB, Beutel
RG, Lawrence JF (eds) Handbook of zoology, vol IV, Arthropoda: Insecta, Teilband 39,
Coleoptera, Beetles, vol 2., Morphology and systematics. Walter de Gruyter, Berlin,
pp 75–103
84. Yeates DK, Wiegmann BM, Courtney GW, Meier R, Lambkin C, Pape T (2007) Phylogeny
and systematics of Diptera: two decades of progress and prospects. Zootaxa 1668:565–590
85. Viviani VR, Hastings JW, Wilson T (2002) Two bioluminescent Diptera: the North
American Orfelia fultoni and the Australian Arachnocampa flava. Similar niche, different
bioluminescence systems. Photochem Photobiol 75:22–27
86. Zompro O, Fritzsche I (1999) Lucihormetica fenestrata n. gen., n. sp., the first record of
luminescence in an orthopteroid insect (Dictyoptera: Blaberidae: Blaberinae: Brachycolini).
Amazoniana 15:211–219
87. Vršanský P, Chorvát D, Fritzsche I, Hain M, Ševěík R (2012) Light-mimicking cockroaches
indicate tertiary origin of recent terrestrial luminescence. Naturwissenschaften 99:739–749
88. Merritt DJ (2013) Standards of evidence for bioluminescence in cockroaches.
Naturwissenschaften 100:697–698
89. Goemans G (2006) The Fulgoridae (Hemiptera, Fulgoromorpha) of Guatemala. In: Cano EB
(ed) Biodiversidad de Guatemala, vol 1. Pub Univ del Vall de Guatemala, Guatemala,
pp 337–344
90. Wood KV (1995) The chemical mechanism and evolutionary development of beetle
bioluminescence. Photochem Photobiol 62:662–673
91. Bocakova M, Bocak L, Hunt T, Teraväinen M, Vogler AP (2007) Molecular phylogenetics
of Elateriformia (Coleoptera): evolution of bioluminescence and neoteny. Cladistics
23:477–496
92. Timmermans MJTN, Vogler AP (2012) Phylogenetically informative rearrangements in
mitochondrial genomes of Coleoptera, and monophyly of aquatic Elateriform beetles
(Dryopoidea). Mol Phylogenet Evol 63:299–304
Eco-Evo Bioluminescence on Land and in the Sea 33
117. Nakamura H, Musicki B, Kishi Y, Shimomura O (1988) Structure of the light emitter in krill
(Euphausia pacifica) bioluminescence. J Am Chem Soc 110:2683–2685
118. Nakamura H, Oba Y, Murai A (1993) Synthesis and absolute configuration of the ozonolysis
product of krill fluorescent compound F. Tetrahedron Lett 34:2779–2782
119. Dunlap JC, Hastings JW, Shimomura O (1980) Crossreactivity between the light-emitting
systems of distantly related organisms: novel type of light-emitting compound. Proc Natl
Acad Sci USA 77:1394–1397
120. Herring PJ (1985) Bioluminescence in the Crustacea. J Crustacean Biol 5:557–573
121. Morin JG (2011) Based on a review of the data, use of the term ‘cypridinid’ solves the
Cypridina/Vargula dilemma for naming the constituents of the luminescent system of
ostracods in the family Cypridinidae. Luminescence 26:1–4
122. Oba Y, Tsuduki H, Kato S, Ojika M, Inouye S (2004) Identification of the luciferin-
luciferase system and quantification of coelenterazine by mass spectrometry in the deep-sea
luminous ostracod Conchoecia pseudodiscophora. ChemBioChem 5:1495–1499
123. Haneda Y, Johnson FH, Shimomura O (1966) The origin of luciferin in the luminous ducts
of Parapriacanthus ransonneti, Pempheris klunzingeri, and Apogon ellioti. In: Johnson FH,
Haneda Y (eds) Bioluminescence in progress. Princeton University Press, Massachusetts
124. Tsuji FI, Haneda Y, Lynch RV III, Sugiyama N (1971) Luminescence cross-reactions of
Porichthys luciferin and theories on the origin of luciferin in some shallow-water fishes.
Comp Biochem Physiol 40A:163–179
125. Haneda Y, Johnson FH, Sie EH-C (1958) Luciferin and luciferase extracts of a fish, Apogon
marginatus, and their luminescent cross-reactions with those of a crustacean, Cypridina
hilgendorfii. Biol Bull 115:336
126. Haneda Y, Johnson FH (1958) The luciferin-luciferase reaction in a fish, Parapriacanthus
beryciformis, of newly discovered luminescence. Proc Natl Acad Sci USA 44:127–129
127. Haneda Y, Tsuji FI, Sugiyama N (1969) Luminescent systems in apogonid fishes from the
Philippines. Science 165:188–190
128. Haneda Y, Tsuji FI, Sugiyama N (1969) Newly observed luminescence in apogonid fishes
from the Philippines. Sci Rept Yokosuka City Mus 15:1–9 + 2 plt
129. Tominaga Y (1963) A revision of the fishes of the family Pempheridae of Japan. J Fac Sci
Univ Tokyo, Section IV Zoology 10:269–290
130. Mooi RD, Jubb RN (1996) Descriptions of two new species of the genus Pempheris (Pisces:
Pempherididae) from Australia, with a provisional key to Australian species. Rec Australian
Mus 48:117–130
131. Johnson FH, Sugiyama N, Shimomura O, Saiga Y, Haneda Y (1961) Crystalline luciferin
from a luminescent fish, Parapriacanthus beryciformes. Proc Natl Acad Sci USA
47:486–489
132. Tsuji FI, Barnes AT, Case JF (1972) Bioluminescence in the marine teleost, Porichthys
notatus, and its induction in a non-luminous form by Cypridina (ostracod) luciferin. Nature
237:515–516
133. Oba Y, Kato S, Ojika M, Inouye S (2002) Biosynthesis of luciferin in the sea firefly,
Cypridina hilgendorfii: L-tryptophan is a component in Cypridina luciferin. Tetrahedron
Lett 43:2389–2392
134. Kato S, Oba Y, Ojika M, Inouye S (2004) Identification of the biosynthetic units of
Cypridina luciferin in Cypridina (Vargula) hilgendorfii by LC/ESI-TOF-MS. Tetrahedron
60:11427–11434
135. Kato S, Oba Y, Ojika M, Inouye S (2006) Stereoselective incorporation of isoleucine into
Cypridina luciferin in Cypridina hilgendorfii (Vargula hilgendorfii). Biosci Biotechnol
Biochem 70:1528–1532
136. Kato S, Oba Y, Ojika M, Inouye S (2007) Biosynthesis of Cypridina luciferin in Cypridina
noctiluca. Heterocycles 72:673–676
137. Goto T, Fukatsu H (1969) Cypridina bioluminescence VII. Chemiluminescence in micelle
solutions: a model system for Cypridina bioluminescence. Tetrahedron Lett 10:4299–4302
Eco-Evo Bioluminescence on Land and in the Sea 35
138. Beebe W (1937) Preliminary list of Bermuda deep-sea fish. Based on the collections from
fifteen hundred metre-net hauls, made in an eight-mile circle South and Nonsuch Island.
Bermuda. Zoologica NY 22:197–208
139. Dunlap PV, Urbanczyk H (2013) Luminous bacteria. In: Rosenberg E (ed) The prokaryotes:
prokaryotic physiology and biochemistry. Springer, Berlin, pp 495–528
140. Herring PJ (2002) Marine microlights: the luminous marine bacteria. Microbiol Today
29:174–176
141. Dunlap PV, Ast JC, Kimura S, Fukui A, Yoshino T, Endo H (2007) Phylogenetic analysis of
host-symbiont specificity and codivergence in bioluminescent symbioses. Cladistics
23:507–532
142. Urbanczyk H, Ast JC, Higgins MJ, Carson J, Dunlap PV (2007) Reclassification of Vibrio
fischeri, Vibrio logei, Vibrio salmonicida and Vibrio wodanis as Aliivibrio fischeri gen. nov.,
comb. nov., Aliivibrio logei comb. nov., Aliivibrio salmonicida comb. nov. and Aliivibrio
wodanis comb. nov. Int J Syst Evol Microbiol 57:2823–2829
143. Meyer-Rochow VB (1976) Loss of bioluminescence in Anomalops katoptron due to
starvation. Experientia 32:1175–1176
144. Haddock SHD, Case JF (1994) A bioluminescent chaetognath. Nature 367:225–226
145. Campbell AK, Herring PJ (1990) Imidazolopyrazine bioluminescence in copepods and
other marine organisms. Mar Biol 104:219–225
146. Miya M, Nemoto T (1986) Reproduction, growth and vertical distribution of the
mesopelagic fish Cyclothone pseudopallida (family Gonostomatidae). In: Uyeno T, Arai
R, Taniuchi T, Matsuura K (eds) Proceedings of the second international conference on the
Indo-Pacific fishes. The Ichthyological Society of Japan, Tokyo, pp 830–837
147. Mallefet J, Shimomura O (1995) Presence of coelenterazine in mesopelagic fishes from the
Strait of Messina. Mar Biol 124:381–385
148. Frank TM, Widder EA, Latz MI, Case JF (1984) Dietary maintenance of bioluminescence in
a deep-sea mysid. J Exp Biol 109:385–389
149. Haddock SHD, Rivers TJ, Robison BH (2001) Can coelenterates make coelenterazine?
Dietary requirement for luciferin in cnidarian bioluminescence. Proc Natl Acad Sci USA
98:11148–11151
150. Thomson CM, Herring PJ, Campbell AK (1995) Evidence for de novo biosynthesis of
coelenterazine in the bioluminescent midwater shrimp, Systellaspis debilis. J Mar Biol Ass
UK 75:165–171
151. Buskey EJ, Stearns DE (1991) The effects of starvation on bioluminescence potential and
egg release of the copepod Metridia longa. J Plankton Res 13:885–893
152. Oba Y, Kato S, Ojika M, Inouye S (2009) Biosynthesis of coelenterazine in the deep-sea
copepod, Metridia pacifica. Biochem Biophys Res Commun 390:684–688
153. Mauchline J (1998) The biology of calanoid copepods: advances in marine biology, vol 33.
Academic Press, San Diego
154. Padmavati G, Ikeda T, Yamaguchi A (2004) Life cycle, population structure and vertical
distribution of Metridia spp. (Copepoda: Calanoida) in the Oyashio region (NW Pacific
Ocean). Mar Ecol Prog Ser 270:181–198
155. Anderson OR (1980) Radiolaria. Springer, New York
156. Meyer-Rochow VB (1986) Luminescent Copepoda of the genus Metridia with special
reference to the Antarctic Metridia gerlachei. New Zld Antarc Rec 7:1–8
157. Hirakawa K, Imamura A (1993) Seasonal abundance and life history of Metridia pacifica
(Copepoda: Calanoida) in Toyama Bay, Southern Japan Sea. Bull Plankton Soc Japan
40:41–54
158. Hayashi S, Hirakawa K (1997) Diet composition of the firefly squid, Watasenia scintillans,
from Toyama Bay, Southern Japan Sea. Bull Japan Sea Natl Fish Res Inst 47:57–66 (in
Japanese with English title and abstract)
159. Campbell AK (2012) Darwin shines light on the evolution of bioluminescence.
Luminescence 27:447–449
36 Y. Oba and D. T. Schultz
160. Johnsen S, Franck TM, Haddock SHD, Widder EA, Messing CG (2012) Light and vision in
the deep-sea benthos: I. Bioluminescence at 500–1000 m depth in the Bahamian Islands.
J Exp Biol 215:3335–3343
161. Ross DM (1959) The sea anemone (Calliactis parasitica) and the hermit crab (Eupagurus
bernhardus). Nature 4693:1161–1162
162. Okamura O (1970) Studies on the macrouroid fishes of Japan: morphology, ecology and
phylogeny. Rept Usa Mar Biol Station 17:1–179 + 5 plt
163. Kanda S (1935) Fireflies (Hotaru), Nippon Hakko Seibutsu Kenkyu Kai, Tokyo (in
Japanese) (reprinted edition, 1981, Scientist Inc, Tokyo)
164. Burkenroad MD (1943) A possible function of bioluminescence. J Mar Res 2:161–164
165. Meyer-Rochow VB (2007) Glowworms: a review of Arachnocampa spp. and kin.
Luminescence 22:251–265
166. Buck J (1988) Synchronous rhythmic flashing of fireflies. Part II. Q Rev Biol 63:265–289
167. Ohba N (1999) Synchronous flashing of the firefly, Pteroptyx effulgens, in Papua New
Guinea. Sci Rept Yokosuka City Mus 46:33–40 (in Japanese with English title and abstract)
168. Anctil M, Case JF (1977) The caudal luminous organs of lanternfishes: general innervation
and ultrastructure. Am J Anat 149:1–22
169. Widder EA (1998) A predatory use of counter illumination by the squaloid shark, Isistius
brasiliensis. Env Biol Fish 53:267–273
170. Abrahams MV, Townsend LD (1993) Bioluminescence in dinoflagellates: a test of the
burglar alarm hypothesis. Ecology 74:258–260
171. Oliveira AG, Stevani CV (2009) The enzymatic nature of fungal bioluminescence.
Photochem Photobiol Sci 8:1416–1421
172. Hebert PDN, Cywinska A, Ball SL, deWaard JR (2003) Biological identifications through
DNA barcodes. Proc R Soc Lond B 270:313–321
173. Branham MA (2010) Lampyridae Latreille, 1817. In: Leschen RAB, Beutel RG, Lawrence
JF (eds) Handbook of zoology, vol IV, Arthropoda: Insecta, Teilband 39, Coleoptera,
Beetles, vol 2., Morphology and systematics. Walter de Gruyter, Berlin, pp 141–149
174. Kawashima I, Lawrence JF, Branham MA (2010) Rhagophthalmidae Olivier, 1907. In:
Leschen RAB, Beutel RG, Lawrence JF (eds) Handbook of zoology, vol IV, Arthropoda:
Insecta, Teilband 39, Coleoptera, Beetles, vol 2., Morphology and systematics. Walter de
Gruyter, Berlin, pp 135–140
Biochemistry and Genetics of Bacterial
Bioluminescence
Paul Dunlap
Keywords Bioluminescence Bacterial luciferase Aliivibrio Photobacterium
Vibrio lux genes
Abbreviations
acyl-HSL Acyl-homoserine lactone
FMNH2 Reduced flavin mononucleotide
Kb Kilobases (thousand nucleotides)
kD Kilodaltons
P. Dunlap (&)
Department of Ecology and Evolutionary Biology, University of Michigan,
Ann Arbor, MI 48109-1048, USA
e-mail: pvdunlap@umich.edu
Contents
1 Introduction.......................................................................................................................... 38
2 Species of Luminous Bacteria ............................................................................................ 39
3 Biochemistry of Bacterial Luminescence........................................................................... 42
4 Bacterial lux Genes ............................................................................................................. 43
5 Regulation of lux Operon Expression ................................................................................ 47
6 Origin and Function of Luminescence in Bacteria ............................................................ 51
7 Outlook ................................................................................................................................ 53
References.................................................................................................................................. 53
1 Introduction
Fig. 1 Bacterial luminescence. Colonies of P. mandapamensis from the light organ of the
cardinalfish Siphamia tubifer (Perciformes: Apogonidae) are shown growing on a nutrient
seawater agar plate. The plate was photographed in room light (left) and (the same plate) in the
dark by the light produced by the bacteria (right). From Dunlap et al. [46]
Thirty or more species of bacteria have strains that make light visible to the eye
(Fig. 1) or that at least carry genes for luminescence, the lux genes (Table 1). These
bacteria, which are all Gram-negative, group phylogenetically as members of three
families of Gammaproteobacteria: Vibrionaceae (Aliivibrio, Photobacterium,
Vibrio), Enterobacteriaceae (Photorhabdus), and Shewanellaceae (Shewanella). It
should be noted, however, that most Vibrionaceae species are not luminous and
apparently lack lux genes and that only the few listed species in Enterobacteriaceae
and Shewanellaceae are known to be luminous. Also, many of the species listed
have nonluminous strains that may or may not lack lux genes [4, 19, 38, 60, 204].
The low number of luminous species, relative to the many described species in
Vibrionaceae, is consistent with loss of the lux genes over evolutionary time from
the ancestors of many lineages within the family. Furthermore, nonluminous vari-
ants apparently can arise readily through the loss of one of more core genes of the
lux operon, luxCDABEG (e.g., [204]). It should be noted here also that several
species previously classified variously as members of Photobacterium or Vibrio
(i.e., fischeri, salmonicida, logei, and wodanis) have been reclassified as members of
40 P. Dunlap
Table 1 (continued)
Species Sources Selected references
Photorhabdus
asymbiotica Human skin lesions [53, 58, 99, 149, 200]
luminescens Insect larvae infected with heterorhabditid [21, 36, 58, 181]
nematodes
temperata Insect larvae infected with heterorhabditid [58, 181]
nematodes
a
Ability of this species to luminescence has not been confirmed; the single strain reported as
luminous [153] might not be available
1.1 Mb, respectively, are massively reduced compared to the genomes of other
members of Vibrionacaeae, which typically are 4.5–6.0 Mb, with many metabolic
and regulatory genes lost. Genome reduction and gene loss apparently account for
the inability of these bacteria to grow in laboratory culture [86–88]. For the deep-sea
anglerfish, no additional information is available at this time on the species-level
phylogenetic placement of their symbiotic luminous bacteria.
Despite much recent progress in clarifying the taxonomy of luminous bacteria
(Table 1), the ecological incidence and species-level diversity of luminous bac-
teria remain incompletely defined. One problem is that the luminescence pheno-
type can be lost; strains luminous on primary isolation often become dim or dark
in laboratory culture [3, 134, 171]. Another is that some species that grow well in
laboratory culture at room temperature, i.e., Aliivibrio logei and Shewanella
hanedai, typically produce light visible to the eye only when grown at cooler
temperatures. In some cases, i.e., luminous bacteria infecting crustaceans; [67] and
strains of Aliivibrio fischeri symbiotic with the Hawaiian sepiolid squid, Euprymna
scolopes [22], the bacteria produce a high level of light in their natural habitat but
produce little or no light when grown in laboratory culture. A further complication
is that strains of some bacteria, such V. cholerae, are known to carry lux genes but
apparently do not express them (e.g., [93, 148, 154, 215]). In addition, bacteria
identified as related to Vibrio harveyi and Vibrio cincinnatiensis carry the lux
genes but have been found to have lux gene mutations that result in a dark phe-
notype [143]. Although the phylogenetically scattered incidence of bacteria with
lux genes in Vibrionaceae presumably relates to different ecologies of the different
species, it is not obvious how having and expressing lux genes contributes to the
lifestyle of most luminous bacteria; there are no obvious ecological differences
between luminous and nonluminous species except in the case of those species that
are bioluminescent symbionts of bacterially luminous fish and squid. On-going and
future studies that examine the incidence, lux gene content, and phylogenetic
placement of luminous bacteria will undoubtedly expand understanding of the
species diversity and ecology of bacteria able to produce light.
Along with bacterial luciferase, the substrates, FMNH2 and long-chain fatty
aldehyde, are specific to the bacterial luminescence reaction; bioluminescent
eukaryotes employ different chemistries and luciferases that are not homologous at
the protein or gene sequence levels to bacterial luciferase [76]. In the luminescence
reaction, binding of FMNH2 by the enzyme is followed by interaction with O2 to
form a flavin-4a-hydroperoxide. Association of this complex with aldehyde forms
a highly stable intermediate, the slow decay of which results in oxidation of the
FMNH2 and aldehyde substrates and the emission of light [76, 79]. Quantum yield
for the reaction has been estimated at 0.1–0.2 photons. The reaction is highly
specific for FMNH2, and the aldehyde substrate in vivo is likely to be tetradecanal.
FMNH2 is provided by the activity of an NADH:FMN oxidoreductase (flavin
reductase), which taps reductant from NADH generated in cellular metabolism, for
example, glycolysis and the citric acid cycle. Transfer of reductant from FMNH2
to luciferase occurs by free diffusion. Synthesis of the long-chain aldehyde is
catalyzed by a fatty-acid reductase complex composed of three polypeptides,
an NADPH-dependent acyl protein reductase (r, 54 kDa), an acyl transferase
(t, 33 kDa), and an ATP-dependent synthetase (s, 42 kDa). The complex has a
stoichiometry of r4s4t2–4, and its activity is essential for the production of light in
the absence of exogenously added aldehyde [79, 121, 183, 201]. Luciferases from
different species of luminous bacteria exhibit substantial amino acid residue and
nucleotide sequence identity [45, 122], consistent with a common evolutionary
origin of luminescence in bacteria.
The lux operon, luxCDABEG, contains the genes necessary for light production in
bacteria (Fig. 2). The luxA and luxB genes code for the a and b subunits of
bacterial luciferase, respectively, luxC, luxD, and luxE genes, respectively, code
for the r, s, and t polypeptides of the fatty-acid reductase complex that synthesizes
and recycles aldehyde substrate for luciferase, and luxG, codes for flavin reductase
[112, 122, 141, 178, 183]. The absence of luxG from the lux operons of Photor-
habdus luminescens and newly characterized species of Ca. Photodesmus (Fig. 2)
apparently is compensated for by the activity of a flavin reductase activity coded
for by an Escherichia coli fre-like gene. Homologues of the fre-like gene are found
in various luminous bacteria [212–214].
An additional gene, luxF, which codes for a nonfluorescent flavoprotein, is
present in the lux operons of Photobacterium, between luxB and luxE (Fig. 2). The
LuxF protein might function in the luminescence system by scavenging an
inhibitory side product of the luciferase reaction [130], but it is not necessary for
light production, even in those Photobacterium species that normally carry this
gene [93]. The luxF gene apparently has been secondarily lost in Photobacterium
leiognathi [5].
44 P. Dunlap
Fig. 2 Bacterial luminescence (lux) genes. Shown are the gene content and gene order of lux
operons for those bacteria for which complete lux operon sequence data are available. Contiguous
genes of the lux operons are aligned to highlight commonalities and differences. Four distinct
types of lux operons are evident based on commonalities of gene content, organization, and
sequence similarity: (1) Aliivibrio/Shewanella type, with luxI/luxR regulatory genes; (2)
Photobacterium type, with ribEBHA genes forming a lux-rib operon; (3) Vibrio/Candidatus
Photodesmus type, without linked regulatory genes; and (4) Photorhabdus type, composed of just
five core lux genes, luxCDABE. Additional species of luminous bacteria are listed in Table 1. See
text for details and for information on accessory genes
Fig. 3 Quorum-sensing regulatory circuitry in Vibrio campbellii (Vibrio harveyi). The expression
of lux operon, and of other quorum-sensing–regulated genes, in V. campbellii (previously classified
as V. harveyi; [107] is coordinated by three chemically distinct autoinducers, HAI-1, AI-2Vh, and
CAI-1, that modulate the phosphorylation state of luxU. The synthesis of each autoinducer is
catalyzed by a different protein, LuxM, LuxS, and CqsA, and each is recognized by a different
cytoplasmic membrane-associated two-component histidine–kinase receptor, LuxN, LuxPQ, and
CqsS, respectively. Low concentrations of the autoinducers lead to phosphorylation of LuxO and
via quorum–regulatory RNAs to the destabilization of the luxRVh transcript, thereby blocking lux
operon transcriptional activation by LuxRVh. High concentrations of the autoinducers reverse the
phosphorylation cascade, allowing formation of LuxRVh and activation of lux operon transcription.
Arrows indicate positive interactions or transcriptional activation, whereas bars indicate negative
interactions or blocking of transcription. See the text for details and references. Redrawn from [185]
differences, however, several commonalities to the two systems have been identified
[44]. Recent publications provide additional details on quorum sensing in these
bacteria as well as in nonluminous bacteria [14, 44, 155].
In V. campbellii, lux operon expression is controlled by a multicomponent
phosphorylation/dephosphorylation cascade (Fig. 3). Three chemically distinct
autoinducers are involved: 3-hydroxybutanoyl-HSL (harveyi autoinducer-1,
Biochemistry and Genetics of Bacterial Bioluminescence 49
Fig. 4 Quorum-sensing control of luminescence in A. fischeri. The expression of the lux operon,
and of other quorum-sensing-regulated genes, in A. fischeri is mediated primarily by the
concentration of AI-1, which forms a complex with LuxRAf. Synthesis of AI-1 is dependent on
LuxI, and the AI-1/LuxRAf complex activates luxICDABEG transcription. Together with cAMP,
the CRP protein activates expression from the luxRAf promoter, increasing synthesis of LuxRAf
and potentiating the system to be induced once sufficient AI-1 has accumulated. The presence of
luxI, coding for AI-1 synthase, as part of the lux operon, leads to increased expression from the
lux operon promoter, stimulating AI-1 synthesis in an autocatalytic, positive feedback manner;
the result is a rapid and strong induction of luciferase synthesis once a threshold concentration of
AI-1 is attained. A second autoinducer, AI-2Af interacts with LuxRAf, interfering with the
interaction between AI-1 and LuxRAf. The hypothesized AI-2Af/LuxRAf complex is thought to be
transcriptionally less effective and therefore to function to delay the onset of AI-1/LuxRAf
activation of luxICDABEG transcription. See the text for details and references
The conserved gene content and gene order of the lux operon in bacteria, lux-
CDABEG (Fig. 2), and the high levels of lux gene and Lux protein amino acid
sequence identities among luminous bacteria (e.g., [122]) indicate that all known
bacterial lux operons derive from a single common ancestor. The congruence of
phylogenies based on lux genes and other protein coding genes (and the 16S rRNA
gene) [189] indicates that bacterial luminescence arose within the Vibrionaceae
lineage and mostly likely in the ancestor that gave rise to Aliivibrio, Photobac-
terium, and Vibrio.
The homology of bacterial luciferase to long-chain alkane monooxygenases
[104] suggests that the light-emitting enzyme arose from this family of proteins,
even though none of the other enzymes in the family emit light [76]. With some
light emission, bacteria luciferase might have evolved under ecological selection,
according to the following scenario [168]. A flavoprotein catalyzing fatty acid
a-oxidation reactions with low chemiluminescent quantum yields is postulated to
have mutated under hypoxic conditions to accept FMNH2 as the flavin cofactor,
generating a fortuitously high fluorescence yield, termed ‘‘protobioluminescence,’’
via the 4a-hydroxy-FMNH product [168]. This flavin dependent, aldehyde-oxi-
dizing protoluciferase produced sufficient light, and with an appropriate emission
spectrum, to be detected by phototactic organisms. Ingestion by visually cueing
animals of particles colonized and made luminous by these early luminous bacteria
presumably enhanced their reproduction by bringing them into the animal’s
nutrient-rich digestive system, ensuring the emitter’s survival and thereby possibly
leading to selection for more intense light output [199]. It is possible that early
evolutionary steps leading to protoluciferase involved oxygen detoxification
activity that permitted early anaerobic organisms to survive an increasingly aer-
obic environment [120, 156]. An alternative hypothesis for the evolution of bac-
terial luciferase, involvement in DNA repair [32], has been refuted [194].
A single gene was hypothesized to encode bacterial protoluciferase [144].
Although a single-subunit protoluciferase, monomer or dimer, presumably would
have differed somewhat from the modern-day luciferase a-subunit and therefore
52 P. Dunlap
might have produced light, the inability of either of the extant a or b subunits alone
to produce light in vitro or in vivo [105] argues against the single-gene hypothesis.
Alternatively, bacterial protoluminescence may have arisen following a gene
duplication event that is postulated to have created luxB from luxA [10, 122, 144].
Based on amino acid sequence identities, a tandem duplication of the ancestral luxA
gene, followed by sequence divergence in the duplicated gene, is thought to have
given rise to luxB, leading to the formation of the heterodimeric luciferase present
in extant luminous bacteria. Similarly, a tandem duplication of luxB followed by
loss of approximately 300 nucleotides coding for N-terminus amino acids is thought
to have given rise to luxF in a luminescent ancestor of Photobacterium; this gene
apparently was later secondarily lost in the lineage giving rise to P. leiognathi [5,
10, 122, 144].
The association of the fatty-acid reductase genes, luxCDE, with luxA might
have predated the luxA to luxB gene duplication event. Alternatively, the presence
of ERIC sequences flanking luxA and luxB in Ph. luminescens [123] might mark an
insertion of the luxAB genes into the fatty aldehyde reductase operon during the
evolution of the bacterial luminescence system. The origins and evolution of other
luminescence genes are not well understood [144]. The evolution of the bacterial
luminescence system also involved recruitment of regulatory and other genes to
the lux operon in some species (Fig. 2).
The energetic cost of light production, involving expenditures of carbon,
nitrogen, and ATP in the synthesis of Lux proteins and in their enzymatic activities
[41] indicates that luminescence has physiological or ecological importance for
those bacteria able to express it. In the absence of selection to retain the lux genes,
this cost would lead to loss of function through mutation and gene loss, an evo-
lutionary scenario that probably accounts for the scattered incidence of luminous
strains and species in Vibrionacaeae. As outlined above, bacterial luminescence
might have arisen evolutionarily as a means of coping with oxygen. Consistent
with this possible function, luciferase, as an oxidase, might function as a secondary
respiratory chain that is active when oxygen or iron levels are too low for the
cytoplasmic membrane-associated, ATP-generating electron transport system to
operate. This activity would allow cells expressing luciferase to reoxidize reduced
coenzyme even when oxygen levels are low [75, 76, 78, 135]. Supporting this
view, growth of cytochrome-deficient luminous bacteria is dependent on induction
of luciferase, limitation for iron stimulates light production, low oxygen levels
promote the luminescence of some luminous bacteria, and luciferase synthesis can
be induced under anaerobic conditions [47, 82, 116, 118, 133]. As an alternative or
supplement to the electron transport system, the activity of luciferase in reoxi-
dizing reduced coenzyme could permit cells of luminous bacteria in low oxygen
habitats, such as in animal gut tracts, to continue to transport and metabolize
growth substrates, thereby continuing to gain energy through substrate-level
phosphorylation. Furthermore and consistent with the ecological selection scenario
above, light production presumably facilitates dissemination of luminous bacteria.
The feeding of animals on luminous particles (decaying tissues, fecal pellets, and
moribund animals infected by luminous bacteria), to which they are attracted,
Biochemistry and Genetics of Bacterial Bioluminescence 53
would bring the bacteria into the animal’s nutrient-rich gut tract for additional
rounds of reproduction followed by dispersal [78, 135]. Recent evidence is sup-
portive of this possibility [211]. Alternatively, the function of the bacterial lux
system might be to generate a halotolerant flavodoxin, with light emission an
incidental consequence [95]. Future studies might provide additional support for
these and other proposed functions for luminescence, such as a physiological role
for luciferase activity in bioluminescent symbioses with fish and squid. Proposed
functions will be held to the standard of a demonstrated selective benefit for
luminescence, either physiological or ecological.
7 Outlook
Substantial progress has been made in the past few years in defining the taxonomy
and phylogenetic relationships of luminous bacteria and more fully characterizing
the biochemistry and genetics of bacterial luminescence. Despite this progress,
current understanding of the physiological function and ecological benefit of
luminescence in bacteria remains limited. The long-standing question, ‘‘Why do
bacteria make light?’’ remains essentially unanswered. A more detailed knowledge
of the evolutionary origins and biochemical uniqueness of bacterial luciferase
(e.g., [104]) and a more comprehensive understanding of the phylogenetic distri-
bution of lux genes through whole-genome sequence analysis (e.g., [193]) in the
context of the ecology of these bacteria (e.g., [211]) are likely to provide clues.
Particularly insightful, however, will likely be detailed comparative physiological
analysis of genetically defined mutants (e.g., [116]), an approach that addresses the
core question and provides an experimental foundation for testing specific func-
tional hypotheses.
References
1. Adar YY, Simaan M, Ulitzur S (1992) Formation of the LuxR protein in the Vibrio fischeri
lux system is controlled by HtpR through the GroESL proteins. J Bacteriol 174:7138–7143
2. Adar YY, Ulitzur S (1993) GroESL proteins facilitate binding of externally added inducer
by LuxR protein–containing E. coli cells. J Biolumin Chemilumin 8:261–266
3. Akhurst RJ (1980) Morphological and functional dimorphism in Xenorhabdus spp., bacteria
symbiotically associated with the insect pathogenic nematodes Neoaplectana and
Heterorhabditis. J Gen Microbiol 121:303–309
4. Akhurst RJ, Boemare NE (1986) A non–luminescent strain of Xenorhabdus luminescens.
J Gen Microbiol 132:1917–1922
5. Ast JC, Cleenwerck I, Engelbeen K, Urbanczyk H, Thompson FL, De Vos P, Dunlap PV,
Ast JC, Dunlap PV (2004) Phylogenetic analysis of the lux operon distinguishes two
evolutionarily distinct clades of Photobacterium leiognathi. Arch Microbiol 181:352–361
6. Ast JC, Dunlap PV (2005) Phylogenetic resolution and habitat specificity of members of the
Photobacterium phosphoreum species group. Environ Microbiol 7:1641–1654
54 P. Dunlap
7. Ast JC, Dunlap PV (2007a) Photobacterium kishitanii sp. nov., a luminous marine
bacterium symbiotic with deep–sea fish. Int J Syst Evol Microbiol 57:2073–2078
8. Ast JC, Urbanczyk H, Dunlap PV (2007) Natural merodiploidy of the lux–rib operon
of Photobacterium leiognathi from coastal waters of Honshu, Japan. J Bacteriol
189:6148–6158
9. Ast JC, Urbanczyk H, Dunlap PV (2009) Multi–gene analysis reveals previously
unrecognized phylogenetic diversity in Aliivibrio. Syst Appl Microbiol 32:379–386
10. Baldwin TO, Ziegler MM, Powers DA (1979) Covalent structure of subunits of bacterial
luciferase NH2– terminal sequence demonstrates subunit homology. Proc Natl Acad Sci
USA 76:4887–4889
11. Baldwin TO, Treat ML, Daubner SC (1990) Cloning and expression of the luxY gene from
Vibrio fischeri strain Y-1 in Escherichia coli and complete amino acid sequence of yellow
fluorescent protein. Biochemistry 29:5509–5515
12. Bang SS, Baumann P, Baumann L (1978) Phenotypic characterization of Photobacterium
logei (sp. nov.), a species related to P. fischeri. Curr Microbiol 1:285–288
13. Bassler BL (1999) How bacteria talk to each other: regulation of gene expression by quorum
sensing. Curr Opin Microbiol 2:582–587
14. Bassler B, Miller MB (2013) Quorum sensing. In: Rosenberg E, DeLong EF, Thompson F,
Lory S, Stackebrandt E (eds), The Prokaryotes (4th ed)—Prokaryotic Communities and
Ecophysiology. Springer, Berlin, pp 495–509. doi:10.1007/978-3-642-30123-0_60
15. Bassler BL, Greenberg EP, Stevens AM (1997) Cross-species induction of luminescence in
the quorum sensing bacterium Vibrio harveyi. J Bacteriol 179:4043–4045
16. Bassler BL, Wright M, Showalter RE, Silverman MR (1993) Intercellular signalling in
Vibrio harveyi, sequence and function of genes regulating expression of luminescence.
Molec Microbiol 9:773–786
17. Bassler BL, Wright M, Silverman MR (1994a) Sequence and function of LuxO, a negative
regulator of luminescence in Vibrio harveyi. Molec Microbiol 12:403–412
18. Bassler BL, Wright M, Silverman MR (1994) Multiple signalling systems controlling
expression of luminescence in Vibrio harveyi, sequence and function of genes encoding a
second sensory pathway. Molec Microbiol 13:273–286
19. Baumann P, Baumann L (1981) The marine Gram–negative eubacteria genera
Photobacterium, Beneckea, Alteromonas, Pseudomonas, and Alcaligenes. In: Starr MP,
Stolp H, Trüper HG, Balows A, Schlegel HG (eds) The prokaryotes. Springer, Berlin,
pp 1302–1331
20. Baumann P, Baumann L, Bang SS, Woolkalis MJ (1980) Reevaluation of the taxonomy of
Vibrio, Beneckea, and Photobacterium: abolition of the genus Beneckea. Curr Microbiol
4:127–132
21. Boemare NE, Akhurst RJ, Mourant RG (1993) DNA relatedness between Xenorhabdus spp.
(Enterobacteriaceae), symbiotic bacteria of Entomopathogenic nematodes, and a proposal
to transfer Xenorhabdus luminescens to a new genus, Photorhabdus gen. nov. Int J Syst
Bacteriol 43:249–255
22. Boettcher KJ, Ruby EG (1990) Depressed light emission by symbiotic Vibrio fischeri of the
sepiolid squid Euprymna scolopes. J Bacteriol 17:3701–3706
23. Boisvert H, Chatelain R, Bassot J-M (1967) Étude d’un Photobacterium isolé de l’organe
lumineux des poissons Leiognathidae. Ann Inst Pasteur Paris 112:520–524
24. Boyle R (1668) Experiments concerning the relation between light and air in shining wood
and fish. Philos Trans 2:581–600
25. Budsberg KJ, Wimpee CF, Braddock JF (2003) Isolation and identification of
Photobacterium phosphoreum from an unexpected niche migrating salmon. Appl Environ
Microbiol 69:6938–6942
26. Callahan SM, Dunlap PV (2000) LuxR– and acylhomoserine– lactone–controlled non–lux
genes define a quorum–sensing regulon in Vibrio fischeri. J Bacteriol 182:2811–2822
27. Cao J-G, Meighen EA (1989) Purification and structural identification of an autoinducer for
the luminescence system of Vibrio harveyi. J Biol Chem 264:21670–21676
Biochemistry and Genetics of Bacterial Bioluminescence 55
28. Chatterjee J, Miyamoto CM, Meighen EA (1996) Autoregulation of luxR the Vibrio harveyi
lux–operon activator functions as a repressor. Molec Microbiol 20:415–425
29. Chatterjee J, Miyamoto CM, Zouzoulas A, Lang BF, Skouris N, Meighen EA (2002) MetR
and CRP bind to the Vibrio harveyi lux promoters and regulate luminescence. Molec
Microbiol 46:101–111
30. Chen P-F, Tu S-C, Hagag N, Wu FY-H, Wu C-W (1985) Isolation and characterization of a
cyclic AMP receptor protein from luminous Vibrio harveyi cells. Arch Biochem Biophys
241:425–431
31. Chen X, Schauder S, Potier N, Dorsselaer AV, Pelczer I, Bassler BL, Hughson FM (2002)
Structural identification of a bacterial quorum–sensing signal containing boron. Nature
415:545–549
32. Czy_z A, Plata K, Wegrzyn G (2003) Stimulation of DNA repair as an evolutionary drive for
bacterial luminescence. Luminescence 18:140–144
33. Daubner SC, Astorga AM, Leisman GB, Baldwin TO (1987) Yellow light emission of
Vibrio fischeri strain Y-1: purification and characterization of the energy-accepting yellow
fluorescent protein. Proc Natl Acad Sci 84:8912–8916
34. Devine JH, Countryman C, Baldwin TO (1988) Nucleotide sequence of the luxR and luxI
genes and structure of the primary regulatory region of the lux regulon of Vibrio fischeri
ATCC 7744. Biochemistry 27:837–842
35. Dolan KM, Greenberg EP (1992) Evidence that GroEL, not r32, is involved in transcription
regulation of the Vibrio fischeri luminescence genes in Escherichia coli. J Bacteriol
174:5132–5135
36. Duchaud E, Rusniok C, Frangeul L, Buchrieser C, Givaudan A, Taourit S, Bocs S,
Boursaux-Eude C, Chandler M, Charles JF, Dassa E, Derose R, Derzelle S, Freyssinet G,
Gaudriault S, Médigue C, Lanois A, Powell K, Siguier P, Vincent R, Wingate V, Zouine M,
Glaser P, Boemare N, Danchin A, Kunst F (2003) The genome sequence of the
entomopathogenic bacterium Photorhabdus luminescens. Nat Biotechnol 21:1307–1313
37. Dunlap PV (1989) Regulation of luminescence by cyclic AMP in cya–like and crp–like
mutants of Vibrio fischeri. J Bacteriol 171:1199–1202
38. Dunlap PV, Ast JC (2005) Genomic and phylogenetic characterization of the luminous
bacteria symbiotic with the deep–sea fish Chlorophthalmus albatrossis (Aulopiformes
Chlorophthalmidae). Appl Environ Microbiol 71:930–939
39. Dunlap PV, Greenberg EP (1985) Control of Vibrio fischeri luminescence gene expression
in Escherichia coli by cyclic AMP and cyclic AMP receptor protein. J Bacteriol 164:45–50
40. Dunlap PV, Greenberg EP (1988) Control of Vibrio fischeri lux gene transcription by a
cyclic AMP receptor protein–LuxR protein regulatory circuit. J Bacteriol 170:4040–4046
41. Dunlap PV, Greenberg EP (1991) Role of intercellular chemical communication in the
Vibrio fischeri—monocentrid fish symbiosis. In: Dworkin M (ed) Microbial Cell–Cell
Interactions. American Society for Microbiology Washington, DC, pp 219–253
42. Dunlap PV, Kuo A (1992) Cell density–dependent modulation of the Vibrio fischeri
luminescence system in the absence of autoinducer and LuxR protein. J Bacteriol
174:2440–2448
43. Dunlap PV, Ray JM (1989) Requirement for autoinducer in transcriptional negative
autoregulation of the Vibrio fischeri luxR gene in Escherichia coli. J Bacteriol 171:3549–3552
44. Dunlap PV, Urbanczyk H (2013) Luminous bacteria. In: Rosenberg E, DeLong EF, Thompson
F, Lory S, Stackebrandt E (eds), The Prokaryotes (4th ed)—Prokaryotic Physiology and
Biochemistry. Springer, Berlin, pp 495–528. doi:10.1007/978-3-642-30141-4_75
45. Dunlap PV, Ast JC, Kimura S, Fukui A, Yoshino T, Endo H (2007) Phylogenetic analysis of
host–symbiont specificity and codivergence in bioluminescent symbioses. Cladistics
23:507–523
46. Dunlap PV, Gould AL, Wittenrich ML, Nakamura M (2012) Symbiosis initiation in the
bacterially luminous sea urchin cardinalfish Siphamia versicolor. J Fish Biol 81:1340–1356
56 P. Dunlap
47. Eberhard A, Hinton JP, Zuck RM (1979) Luminous bacteria synthesize luciferase
anaerobically. Arch Microbiol 121:277–282
48. Eberhard A, Burlingame AL, Eberhard C, Kenyon GL, Nealson KH, Oppenheimer NJ
(1981) Structural identification of autoinducer of Photobacterium fischeri luciferase.
Biochemistry 20:2444–2449
49. Eberhard A, Widrig CA, McBath P, Schineller JB (1986) Analogs of the autoinducer of
bioluminescence in Vibrio fischeri. Arch Microbiol 146:35–40
50. Eberhard A, Longin T, Widrig CA, Stranick SJ (1991) Synthesis of the lux gene autoinducer
in Vibrio fischeri is positively autoregulated. Arch Microbiol 155:294–297
51. Engebrecht J, Silverman M (1984) Identification of genes and gene products necessary for
bacterial bioluminescence. Proc Natl Acad Sci USA 81:4154–4158
52. Engebrecht J, Nealson K, Silverman M (1983) Bacterial bioluminescence, isolation and
genetic analysis of functions from Vibrio fischeri. Cell 32:773–781
53. Farmer JJ, Jorgensen JH, Grimont PAD, Akhurst RJ, Poinar GO, Pierce GV, Smith JA,
Carger GP, Wilson K, Hickman-Brenner FW (1989) Xenorhabdus luminescens (DNA
hybridization group 5) from human clinical specimens. J Clin Microbiol 27:1594–1600
54. Fidopiastis PM, von Boletzky S, Ruby EG (1998) A new niche for Vibrio logei, the
predominant light organ symbiont of squids in the genus Sepiola. J Bacteriol 180:59–64
55. Fidopiastis PM, Sorum H, Ruby EG (1999) Cryptic luminescence in the cold–water fish
pathogen Vibrio salmonicida. Arch Microbiol 171:205–209
56. Fidopiastis PM, Miyamoto CM, Jobling MG, Meighen EG, Ruby EG (2002) LitR, a new
transcriptional activator in Vibrio fischeri, regulates luminescence and symbiotic light organ
colonization. Molec Microbiol 45:131–143
57. Figge MJ, Robertson LA, Ast JC, Dunlap PV (2011) Historical microbiology: revival and
phylogenetic characterization of luminous bacterial cultures of M. W. Beijerinck. FEMS
Microbiol Ecol 78:463–472
58. Fischer–Le Saux M, Viallard V, Brunel B, Normand P, Boemare EN (1999) Polyphasic
classification of the genus Photorhabdus and proposal of new taxa P. luminescens subsp.
luminescens subsp. nov., P. luminescens subsp. akhurstii subsp. nov., P. luminescens subsp.
laumondii subsp. nov., P. temperata sp. nov., P. temperata subsp. temperata subsp. nov.,
and P. asymbiotica sp. nov. Int J Syst Bacteriol 49:1645–1656
59. Fitzgerald JM (1977) Classification of luminous bacteria from the light organ of the
Australian pinecone fish, Cleidopus gloriamaris. Arch Microbiol 112:153–156
60. Forst S, Nealson K (1996) Molecular biology of the symbiotic–pathogenic bacteria
Xenorhabdus spp. and Photorhabdus spp. Microbiol Rev 60:21–43
61. Forst S, Dowds B, Boemare N, Stackebrandt E (1997) Xenorhabdus and Photorhabdus spp.
Bugs that kill bugs. Ann Rev Microbiol 51:47–72
62. Freeman JA, Bassler BL (1999) A genetic analysis of the function of LuxO, a two–component
response regulator involved in quorum sensing in Vibrio harveyi. Molec Microbiol
31:665–677
63. Freeman JA, Bassler BL (1999) Sequence and function of LuxU: a two–component
phosphorelay protein that regulates quorum sensing in Vibrio harveyi. J Bacteriol
191:899–906
64. Freeman JA, Lilley BN, Bassler BL (2000) A genetic analysis of the functions of LuxN: a
two–component hybrid sensor kinase that regulates quorum sensing in Vibrio harveyi.
Molec Microbiol 35:139–149
65. Fukasawa S, Dunlap PV (1986) Identification of luminous bacteria isolated from the light
organ of the squid, Doryteuthis kensaki. J Agric Biol Chem 50:1645–1646
66. Fuqua WC, Winans SC, Greenberg EP (1994) Quorum sensing in bacteria the LuxR–LuxI
family of cell density–responsive transcriptional regulators. J Bacteriol 176:269–275
67. Giard A, Billet A (1889) Observations sur la maladie phosphorescente des Talitres et autres
crustaces. Compt Rend Biol Paris 41:593–597
68. Gilson L, Kuo A, Dunlap PV (1995) AinS and a new family of autoinducer synthesis
proteins. J Bacteriol 177:6946–6951
Biochemistry and Genetics of Bacterial Bioluminescence 57
92. Jensen MJ, Tebo BM, Baumann P, Mandel M, Nealson KH (1980) Characterization of
Alteromonas hanedai (sp. nov.), a nonfermentative luminous species of marine origin. Curr
Microbiol 3:311–315
93. Kaeding AJ, Ast JC, Pearce MM, Urbanczyk H, Kimura S, Endo H, Nakamura M, Dunlap
PV (2007) Phylogenetic diversity and co–symbiosis in the bioluminescent symbioses of
Photobacterium mandapamensis. Appl Environ Microbiol 73:3173–3182
94. Kaplan HB, Greenberg EP (1985) Diffusion of autoinducer is involved in regulation of the
Vibrio fischeri luminescence system. J Bacteriol 163:1210–1214
95. Kasai S (2006) Freshwater bioluminescence in Vibrio albensis (Vibrio cholerae biovar
albensis) NCIMB 41 is caused by a two–nucleotide deletion in luxO. J Biochem 139:471–482
96. Kasai S, Okada K, Hoshino A, Iida T, Honda T (2007) Lateral transfer of the lux gene
cluster. J Biochem Tokyo 141:231–237
97. Kelly RC, Bolitho ME, Higgins DA, Lu W, Ng WL, Jeffrey PD, Rabinowitz JD,
Semmelhack MF, Hughson FM, Bassler BL (2009) The Vibrio cholerae quorum–sensing
autoinducer CAI–1: analysis of the biosynthetic enzyme CqsA. Nat Chem Biol 5:891–895
98. Kuo A, Callahan SM, Dunlap PV (1996) Modulation of luminescence operon expression by
N–octanoyl–homoserine lactone in ainS mutants of Vibrio fischeri. J Bacteriol 178:971–976
99. Kuwata R, Yoshiga T, Yoshida M, Kondo E (2008) Mutualistic association of
Photorhabdus asymbiotica with Japanese heterorhabditid entomopathogenic nematodes.
Microbes Infect 10:734–741
100. Lee CY, Meighen EA (1992) The lux genes in Photobacterium leiognathi are closely linked
with genes corresponding in sequence to riboflavin synthesis genes. Biochem Biophys Res
Commun 186:690–697
101. Lee CY, O’Kane DJ, Meighen EA (1994) Riboflavin synthesis genes are linked with the lux
operon of Photobacterium phosphoreum. J Bacteriol 176:2100–2104
102. Lee J (1993) Lumazine protein and the excitation mechanism in bacterial bioluminescence.
Biophys Chem 48:149–158
103. Lenz DH, Mok KC, Lilley BN, Kulkarni RV, Wingreen NS, Bassler BL (2004) The small
RNA chaperone Hfq and multiple small RNAs control quorum sensing in Vibrio harveyi.
Cell 118:69–82
104. Li L, Liu X, Yang W, Xu W, Xu F, Wang W, Feng L, Bartlam M, Wang L, Rao Z (2008)
Crystal structure of long-chain alkane monooxygenase (LadA) in complex with coenzyme
FMN: unveiling the long-chain alkane hydroxylase. J Mol Biol 376:453–465
105. Li Z, Szittner R, Meighen EA (1993) Subunit interactions and the role of the luxA
polypeptide in controlling thermal stability and catalytic properties in recombinant
luciferase hybrids. Biochim Biophys Acta 1158:137–145
106. Lilley BN, Bassler BL (2000) Regulation of quorum sensing in Vibrio harveyi by LuxO and
sigma–54. Molec Microbiol 36:940–954
107. Lin B, Wang Z, Malanoski AP, O’Grady EA, Wimpee CF, Vuddhakul V, Alves N Jr,
Thompson FL, Gomez-Gil B, Voral GJ (2010) Comparative genomic analyses identify the
Vibrio harveyi genome sequenced strains BAA–1116 and HY01 as Vibrio campbellii.
Environ Microbiol Rep 2:81–89
108. Lin J-W, Chao Y-F, Weng S-F (1993) The lumazine protein–encoding gene in
Photobacterium leiognathi is linked to the lux operon. Gene 126:153–154
109. Lin J-W, Yu K-Y, Chao Y-F, Weng S-F (1995) The lumQ gene is linked to the lumP gene
and the lux operon in Photobacterium leiognathi. Biochem Biophys Res Commun
217:684–695
110. Lin J-W, Chao Y-F, Weng S-F (1996) Nucleotide sequence and functional analysis of the
luxE gene encoding acyl–protein synthetase of the lux operon from Photobacterium
leiognathi. Biochem Biophys Res Commun 228:764–773
111. Lin J-W, Yu K-Y, Chao Y-F, Weng S-F (1996) Regulatory region with putA gene of proline
dehydrogenase that links to the lum and lux operons in Photobacterium leiognathi. Biochem
Biophys Res Commun 219:868–875
Biochemistry and Genetics of Bacterial Bioluminescence 59
112. Lin J-W, Chao Y-F, Weng S-F (1998) Characteristic analysis of the luxG gene encoding the
probable flavin reductase that resides in the lux operon of Photobacterium leiognathi.
Biochem Biophys Res Commun 246:446–452
113. Lin J-W, Chao Y-F, Weng S-F (2001) Riboflavin synthesis genes ribE, ribB, ribH, ribA
reside in the lux operon of Photobacterium leiognathi. Biochem Biophys Res Commun
284:587–595
114. Lin YH, Miyamoto C, Meighen EA (2000) Cloning and functional studies of a luxO
regulator LuxT from Vibrio harveyi. Biochim Biophys Acta 1494:226–235
115. Lunder T, Sørum H, Holstad G, Steigerwalt AG, Mowinckel P, Brenner DJ (2000)
Phenotypic and genotypic characterization of Vibrio viscosus sp. nov. and Vibrio wodanis
sp. nov. isolated from Atlantic salmon (Salmo salar) with ‘winter ulcer’. Int J Syst Evol
Microbiol 50:427–450
116. Makemson JC (1986) Luciferase–dependent oxygen consumption by bioluminescent
vibrios. J Bacteriol 165:461–466
117. Makemson JC, Fulayfil NR, Landry W, Van Ert LM, Wimpee CF, Widder EA, Case JF
(1997) Shewanella woodyi sp. nov., an exclusively respiratory luminous bacterium isolated
from the Alboran Sea. Int J Syst Bacteriol 47:1034–1039
118. Makemson JC, Hastings JW (1982) Iron represses bioluminescence in Vibrio harveyi. Curr
Microbiol 7:181–186
119. Manukhov IV, Khrul’nova SA, Baranova A, Zavilgelsky GB (2011) Comparative analysis
of the lux operons in Aliivibrio logei KCh1 (a Kamchatka Isolate) and Aliivibrio
salmonicida. J Bacteriol 193:3998–4001
120. McElroy WD, Seliger HH (1962) Origin and evolution of bioluminescence. In: Kasha M,
Pullman B (eds) Horizons in biochemistry. Academic Press, New York, pp 91–101
121. Meighen EA (1991) Molecular biology of bacterial bioluminescence. Microbiol Rev
55:123–142
122. Meighen EA, Dunlap PV (1993) Physiological, biochemical and genetic control of bacterial
bioluminescence. Adv Microb Physiol 34:1–67
123. Meighen EA, Szittner RB (1992) Multiple repetitive elements and organization of the lux
operons of luminescent terrestrial bacteria. J Bacteriol 174:5371–5381
124. Miller MB, Bassler BL (2001) Quorum sensing in bacteria. Annu Rev Microbiol
55:165–199
125. Miyamoto CM, Chatterjee J, Swartzman E, Szittner R, Meighen EA (1996) The role of lux
autoinducer in regulating luminescence in Vibrio harveyi control of luxR expression. Molec
Microbiol 19:767–775
126. Miyamoto CM, Dunlap PV, Ruby EG, Meighen EA (2003) LuxO controls luxR expression
in Vibrio harveyi evidence for a common regulatory mechanism in Vibrio. Molec Microbiol
48:537–548
127. Miyamoto CM, Graham AF, Meighen EA (1988) Nucleotide sequence of the luxC gene and
the upstream DNA from the bioluminescent system of Vibrio harveyi. Nucl Acids Res
16:1551–1562
128. Miyamoto CM, Lin YH, Meighen EA (2000) Control of bioluminescence in Vibrio fischeri
by the LuxO signal response regulator Molec. Microbiol 36:594–607
129. Miyashiro T, Wollenberg MS, Cao X, Oehlert D, Ruby EG (2010) A single qrr gene is
necessary and sufficient for LuxO–mediated regulation in Vibrio fischeri. Molec Microbiol
77:1556–1567
130. Moore SA, James MN (1995) Structural refinement of the non–fluorescent flavoprotein from
Photobacterium leiognathi at 160 A resolution. J Mol Biol 249:195–214
131. Müller-Breitkreutz K, Winkler UK (1993) Anaerobic expression of the Vibrio fischeri lux
regulon in E. coli is Fnr–dependent. J Biolumin Chemilumin 8:108
132. Nealson KH (1977) Autoinduction of bacterial luciferase. Occurrence, mechanism and
significance. Arch Microbiol 112:73–79
133. Nealson KH, Hastings JW (1977) Low oxygen is optimal for luciferase synthesis in some
bacteria. Ecological implications. Arch Microbiol 112:9–16
60 P. Dunlap
134. Nealson KH, Hastings JW (1979) Bacterial bioluminescence. Its control and ecological
significance. Microbiol Rev 43:496–518
135. Nealson KH, Hastings JW (1992) The luminous bacteria. In: Balows A, Trüper HG,
Dworkin M, Harder W, Schleifer K-H (eds) The prokaryotes, 2nd edn. Springer, Berlin,
pp 625–639
136. Nealson KH, Platt T, Hastings JW (1970) Cellular control of synthesis and activity of the
bacterial luminescence system. J Bacteriol 104:313–322
137. Nealson KH, Eberhard A, Hastings JW (1972) Catabolite repression of bacterial
bioluminescence, functional implications. Proc Natl Acad Sci USA 69:1073–1076
138. Nealson KH, Wimpee B, Wimpee C (1993) Identification of Vibrio splendidus as a member
of the planktonic luminous bacteria from the Persian Gulf and Kuwait region with luxA
probes. Appl Environ Microbiol 59:2684–2689
139. Nelson EJ, Tunsjø HS, Fidopiastis PM, Sørum H, Ruby EG (2007) A novel lux operon in the
cryptically bioluminescent fish pathogen Vibrio salmonicida is associated with virulence.
Appl Environ Microbiol 73:1825–1833
140. Ng W-L, Bassler BL (2009) Bacterial quorum–sensing network architectures. Ann Rev Gen
43:197–222
141. Nijvipakul S, Wongratana J, Suadee C, Entsch B, Ballou DP, Chaiyen P (2008) LuxG is a
functioning flavin reductase for bacterial luminescence. J Bacteriol 190:1531–1538
142. O’Brien CH, Sizemore RK (1979) Distribution of the luminous bacterium Beneckea harveyi
in a semitropical estuarine environment. Appl Environ Microbiol 38:933–938
143. O’Grady EA, Wimpee CF (2008) Mutations in the lux operon of natural dark mutants in the
genus Vibrio. Appl Environ Microbiol 74:61–66
144. O’Kane DJ, Prasher DC (1992) Evolutionary origins of bacterial bioluminescence. Molec
Microbiol 6:443–449
145. O’Kane DJ, Karle VA, Lee J (1985) Purification of lumazine proteins from Photobacterium
leiognathi and Photobacterium phosphoreum: bioluminescent properties. Biochemistry
24:1461–1467
146. O’Kane DJ, Woodward B, Lee J, Prasher DC (1991) Borrowed proteins in bacterial
bioluminescence. Proc Natl Acad Sci 88:1100–1104
147. Oliver JD, Roberts DM, White VK, Dry MA, Simpson LM (1986) Bioluminescence in a
strain of the human pathogenic bacterium Vibrio vulnificus. Appl Environ Microbiol
52:1209–1211
148. Palmer LM, Colwell RR (1991) Detection of luciferase gene sequence in nonluminescent
Vibrio cholerae by colony hybridization and polymerase chain reaction. Appl Environ
Microbiol 57:1286–1293
149. Peel MM, Alfredson DA, Gerrard JG, Davis JM, Robson JM, McDougall RJ, Scullie BL,
Akhurst RJ (1999) Isolation, identification, and molecular characterization of strains of
Photorhabdus luminescens from infected humans in Australia. J Clin Microbiol
37:3647–3653
150. Petushkov VN, Lee J (1997) Purification and characterization of flavoproteins and
cytochromes from the yellow bioluminescence marine bacterium Vibrio fischeri strain Y1.
Eur J Biochem 245:790–796
151. Petushkov VN, Ketelaars M, Gibson BG, Lee J (1996) Interaction of Photobacterium
leiognathi and Vibrio fischeri Y1 luciferases with fluorescent (antenna) proteins
bioluminescence effects of the aliphatic additive. Biochemistry 35:12086–12093
152. Pflüger E (1875) Ueber die Phosphorescenz verwesender Organismen. Arch ges Physiol
Men Tiere 11:222–263
153. Pujalte MJ, Garay E (1986) Proposal of Vibrio mediterranei sp. nov. A new marine member
of the Genus Vibrio. Int J Syst Bacteriol 36:278–281
154. Ramaiah N, Chun J, Ravel J, Straube WL, Hill RT, Colwell RR (2000) Detection of
luciferase gene sequences in non–luminescent bacteria from the Chesapeake Bay. FEMS
Microbiol Ecol 33:27–34
Biochemistry and Genetics of Bacterial Bioluminescence 61
155. Ramsey MM, Korgaonkar AK, Whiteley M (2009) Quorum sensing in bacteria. In:
Schaechter M Encyclopedia of microbiology, 3rd edn. Academic Press, New York
156. Rees J-F, de Wergifosse B, Noiset O, Dubuisson M, Janssens B, Thompson EM (1998) The
origins of marine bioluminescence. Turning oxygen defense mechanisms into deep–sea
communication tools. J Exp Biol 201:1211–1221
157. Reichelt JL, Baumman P (1973) Taxonomy of the marine, luminous bacteria. Arch
Mikrobiol 94:283–330
158. Reichelt JL, Nealson K, Hastings JW (1977) The specificity of symbiosis pony fish and
luminescent bacteria. Arch Microbiol 112:157–161
159. Robertson LA, Figge MJ, Dunlap PV (2011) Beijerinck and the bioluminescent bacteria—
microbiological experiments in the late 19th and early 20th centuries. FEMS Microbiol Ecol
75:185–194
160. Ruby EG, Morin JG (1979) Luminous enteric bacteria of marine fish a study of their
distribution, densities, and dispersion. Appl Environ Microbiol 38:406–411
161. Ruby EG, Nealson KH (1976) Symbiotic association of Photobacterium fischeri with the
marine luminous fish Monocentris japonica, a model of symbiosis based on bacterial
studies. Biol Bull 141:574–5867
162. Ruby EG, Nealson KH (1977) A luminous bacterium that emits yellow light. Science
196:432–434
163. Ruby EG, Nealson KH (1978) Seasonal changes in the species composition of luminous
bacteria in nearshore seawater. Limnol Oceanogr 23:530–533
164. Ruby EG, Urbanowski M, Campbell J, Dunn A, Faini M, Gunsalus R, Lostroh P, Lupp C,
McCann J, Millikan D, Schaefer A, Stabb E, Stevens A, Visick K, Whistler C, Greenberg
EP (2005) Complete genome sequence of Vibrio fischeri a symbiotic bacterium with
pathogenic congeners. Proc Natl Acad Sci USA 102:3004–3009
165. Sato Y, Shimizu S, Ohtaki A, Noguchi K, Miyatake H, Dohmae N, Sasaki S, Odaka M,
Yohda M (2010) Crystal structures of the Lumazine protein from Photobacterium kishitanii
in complexes with the authentic chromophore, 6,7–dimethyl–8–(1_–D–ribityl) lumazine,
and its analogues, riboflavin and flavin mononucleotide, at high resolution. J Bacteriol
192:127–133
166. Schaefer AL, Val DL, Hanzelka BL, Cronan JE Jr, Greenberg EP (1996) Generation of cell–
to–cell signals in quorum sensing:acyl homoserine lactone synthase activity of a purified
Vibrio fischeri LuxI. Proc Natl Acad Sci USA 93:9505–9509
167. Schauder S, Shokat K, Surette MG, Bassler BL (2001) The LuxS family of bacterial
autoinducers biosynthesis of a novel quorum–sensing signal molecule. Molec Microbiol
41:463–476
168. Seliger HH (1987) The evolution of bioluminescence in bacteria. Photochem Photobiol
45:291–297
169. Shadel GS, Devine JH, Baldwin TO (1990) Control of the lux regulon of Vibrio fischeri.
J Biolumin Chemilumin 5:99–106
170. Showalter RE, Martin MO, Silverman MR (1990) Cloning and nucleotide sequence of luxR,
a regulatory gene controlling bioluminescence in Vibrio harveyi. J Bacteriol 172:2946–2954
171. Silverman M, Martin M, Engebrecht J (1989) Regulation of luminescence in marine
bacteria. In: Hopwood DA, Chater KF (eds) Genetics of bacterial diversity. Academic Press,
London, pp 71–86
172. Small ED, Koka P, Lee J (1980) Lumazine protein from the bioluminescent bacterium
Photobacterium phosphoreum. Purification and characterization. J Biol Chem 255:8804–8810
173. Smith SK, Sutton DC, Fuerst JA, Reichelt JL (1991) Evaluation of the genus Listonella and
reassignment of Listonella damsela (Love et al.) MacDonell and Colwell to the genus
Photobacterium as Photobacterium damsela comb. nov. with an emended description. Int J
Syst Bacteriol 41:529–534
174. Stevens AM, Greenberg EP (1997) Quorum sensing in Vibrio fischeri: essential elements for
activation of the luminescence genes. J Bacteriol 179:557–562
62 P. Dunlap
175. Suadee C, Nijvipakul S, Svasti J, Entsch B, Ballou DP, Chaiyen P (2007) Luciferase from
Vibrio campbellii is more thermostable and binds reduced FMN better than its homologues.
J Biochem 142:539–552
176. Sung ND, Lee CY (2004) Coregulation of lux genes and riboflavin genes in bioluminescent
bacteria of Photobacterium phosphoreum. J Microbiol 42:194–199
177. Surete MG, Miller MB, Bassler BL (1999) Quorum sensing in Escherichia coli, Salmonella
typhimurium, and Vibrio harveyi: a new family of genes responsible for autoinducer
production. Proc Natl Acad Sci USA 96:1639–1644
178. Swartzman E, Kapoor S, Graham AF, Meighen EA (1990) A new Vibrio fischeri lux gene
precedes a bidirectional termination site for the lux operon. J Bacteriol 172:6797–6802
179. Swartzman E, Miyamoto C, Graham A, Meighen EA (1990) Delineation of the
transcriptional boundaries of the lux operon of Vibrio harveyi demonstrates the presence
of two new lux genes. J Biol Chem 265:3513–3517
180. Swartzman E, Silverman M, Meighen EA (1992) The luxR gene product of Vibrio harveyi is
a transcriptional activator of the lux promoter. J Bacteriol 174:7490–7493
181. Tailliez P, Laroui C, Ginibre N, Paule A, Pagès S, Boemare N (2010) Phylogeny of
Photorhabdus and Xenorhabdus based on universally conserved protein–coding sequences
and implications for the taxonomy of these two genera. Proposal of new taxa X.
vietnamensis sp. nov., P. luminescens subsp. caribbeanensis subsp. nov., P. luminescens
subsp. hainanensis subsp. nov., P. temperata subsp. khanii subsp. nov., P. temperata subsp.
tasmaniensis subsp. nov., and the reclassification of P. luminescens subsp. thracensis as P.
temperata subsp. thracensis comb. nov. Int J Syst Evol Microbiol 60:1921–1937
182. Thompson FL, Thompson CC, Li Y, Gomez-Gil B, Vandenberghe J, Hoste B, Swings J
(2003) Vibrio kanaloae sp. nov., Vibrio pomeroyi sp. nov. and Vibrio chagasii sp. nov.,
from sea water and marine animals. Int J Syst Evol Microbiol 53:753–759
183. Tinikul R, Pitsawong W, Sucharitakul J, Nijvipakul S, Ballou DP, Chaiyen P (2013) The
transfer of reduced flavin mononucleotide from LuxG oxidoreductase to luciferase occurs
via free diffusion. Biochemistry 52:6834–6843
184. Tu KC, Bassler BL (2007) Multiple small RNAs act additively to integrate sensory
information and control quorum sensing in Vibrio harveyi. Genes Dev 21:221–233
185. Tu KC, Long T, Svenningsen SL, Wingreen NS, Bassler BL (2010) Negative feedback
loops involving small regulatory RNAs precisely control the Vibrio harveyi quorum–
sensing response. Mol Cell 37:567–579
186. Ulitzur S, Dunlap PV (1995) Regulatory circuitry controlling luminescence autoinduction in
Vibrio fischeri. Photochem Photobiol 62:625–632
187. Ulitzur S, Yashphe J (1975) An adenosine 3’,5’–monophosphate–requiring mutant of the
luminous bacteria Beneckea harveyi. Biochim Biophys Acta 404:321–328
188. Urbanczyk H, Ast JC, Higgins MJ, Carson J, Dunlap PV (2007) Reclassification of Vibrio
fischeri, Vibrio logei, Vibrio salmonicida and Vibrio wodanis as Aliivibrio fischeri gen. nov.,
comb. nov., Aliivibrio logei comb. nov., Aliivibrio salmonicida comb. nov. and Aliivibrio
wodanis comb. nov. Int J Syst Evol Micro 57:2823–2829
189. Urbanczyk H, Ast JC, Kaeding AJ, Oliver JD, Dunlap PV (2008) Phylogenetic analysis of
the incidence of lux gene horizontal transfer in Vibrionaceae. J Bacteriol 190:3494–3504
190. Urbanczyk H, Ast JC, Dunlap PV (2011) Phylogeny, genomics, and symbiosis of
Photobacterium. FEMS Microbiol Rev 35:324–342
191. Urbanczyk H, Ogura Y, Hendry TA, Gould AL, Kiwaki N, Atkinson JT, Hayashi T, Dunlap
PV (2011) Genome Sequence of Photobacterium mandapamensis svers.1.1, the
bioluminescent symbiont of the cardinal fish Siphamia versicolor. J Bacteriol 193:3144–3145
192. Urbanczyk H, Furukawa T, Yamamoto Y, Dunlap PV (2012) Natural replacement of the
vertically inherited lux-rib genes of Photobacterium aquimaris by horizontally acquired
homologs. Environ Microbiol Rep 4:412–416
Biochemistry and Genetics of Bacterial Bioluminescence 63
193. Urbanczyk H, Ogura Y, Hayashi T (2013) Taxonomic revision of Harveyi clade bacteria
(family Vibrionaceae) base don analysis of whole genome sequences. Int J Syst Evol
Microbiol 63:2742–2751
194. Walker EL, Bose JL, Stabb EV (2006) Photolyase confers resistance to UV light but does
not contribute to the symbiotic benefit of bioluminescence in Vibrio fischeri ES114. Appl
Environ Microbiol 72:6600–6606
195. Waterfield NR, Ciche T, Clarke D (2009) Photorhabdus and a host of hosts. Ann Rev
Microbiol 63:557–574
196. Waters CM, Bassler BL (2005) Quorum sensing cell–to–cell communication in bacteria.
Ann Rev Cell Devel Biol 21:319–346
197. Waters CM, Bassler BL (2006) The Vibrio harveyi quorum-sensing system uses shared
regulatory components to discriminate between multiple autoinducers. Genes Dev
20:2754–2767
198. Wei Y, Perez LJ, Ng WL, Semmelhack MF, Bassler BL (2011) Mechanism of Vibrio
cholerae autoinducer–1 biosynthesis. ACS Chem Biol 6:356–365
199. Widder EA (2010) Bioluminescence in the ocean origins of biological, chemical, and
ecological diversity. Science 328:704–708
200. Wilkinson P, Waterfield NR, Crossman L, Corton C, Sanchez-Contreras M, Vlisidou I,
Barron A, Bignell A, Clark L, Ormond D, Mayho M, Bason N, Smith F, Simmonds M,
Churcher C, Harris D, Thompson NR, Quail M, Parkhill J, Ffrench-Constant RH (2009)
Comparative genomics of the emerging human pathogen Photorhabdus asymbiotica with
the insect pathogen Photorhabdus luminescens. BMC Genom 10:302
201. Wilson T, Hastings JW (1998) Bioluminescence. Ann Rev Cell Devel Biol 14:197–230
202. Wimpee CF, Nadeau T-L, Nealson KH (1991) Development of species–specific
hybridization probes for marine luminous bacteria by using in vitro DNA amplification.
Appl Environ Microbiol 57:1319–1324
203. Wolfe CJ, Haygood MG (1991) Restriction fragment length polymorphism analysis reveals
high levels of genetic divergence among the light organ symbionts of flashlight fish. Biol
Bull 181:135–143
204. Wollenberg MS, Preheim SP, Polz MF, Ruby EG (2012) Polyphyly of non–bioluminescent
Vibrio fischeri sharing a lux–locus deletion. Environ Microbiol 14:655–668
205. Yang Y, Yeh LP, Cao Y, Baumann L, Baumann P, Tang JS-E, Beaman B (1983)
Characterization of marine luminous bacteria isolated off the coast of China and description
of Vibrio orientalis sp. nov. Curr Microbiol 8:95–100
206. Yetinson T, Shilo M (1979) Seasonal and geographic distribution of luminous bacteria in
the eastern Mediterranean Sea and the Gulf of Elat. Appl Environ Microbiol 37:1230–1238
207. Yoshizawa S, Wada M, Kita-Tsukamoto K, Ikemoto E, Yokota A, Kogure K (2009) Vibrio
azureus sp. nov., a luminous marine bacterium isolated from seawater. Int J Syst Evol
Microbiol 59:1645–1649
208. Yoshizawa S, Wada M, Kita-Tsukamoto K, Yokota A, Kogure K (2009) Photobacterium
aquimaris sp. nov., a luminous marine bacterium isolated from seawater. Int J Syst Evol
Microbiol 59:1438–1442
209. Yoshizawa S, Karatani H, Wada M, Yokota A, Kogure K (2010) Aliivibrio sifiae sp. nov.,
luminous marine bacteria isolated from seawater. J Gen Appl Microbiol 56:508–518
210. Yoshizawa S, Wada M, Yokota A, Kogure K (2010) Vibrio sagamiensis sp. nov., luminous
marine bacteria isolated from seawater. J Gen Appl Microbiol 56:499–507
211. Zarubin M, Belkin S, Ionescu M, Genin A (2012) Bacterial bioluminescence as a lure for
marine zooplankton and fish. Proc Natl Acad Sci USA 109:853–857
212. Zenno H, Saigo K (1994) Identification of the genes encoding NAD(P)H–flavin
oxidoreductases that are similar in sequence to Escherichiacoli Fre in four species of
luminous bacteria Photorhabdus luminescens, Vibrio fischeri, Vibrio harveyi, and Vibrio
orientalis. J Bacteriol 176:3544–3551
213. Zenno H, Inouye S, Saigo K (1992) Does the luxG gene in luminous bacteria code for an
NAD(P)H–FMN oxidoreductase? Genetics (Life Sci Adv) 11:85–91
64 P. Dunlap
214. Zenno H, Saigo K, Kanoh H, Inouye S (1994) Identification of the gene encoding the major
NAD(P)H–flavin oxidoreductase of the bioluminescent bacterium Vibrio fischeri ATCC
7744. J Bacteriol 176:3536–3543
215. Zo YG, Chokesajjawatee N, Grim C, Arakawa E, Watanabe H, Colwell RR (2009)
Diversity and seasonality of bioluminescent Vibrio cholerae populations in Chesapeake
Bay. Appl Environ Microbiol 75:135–146
Part II
Applications of Bioluminescence in
Environment and Security
Application of Enzyme Bioluminescence
in Ecology
Contents
1 Introduction.......................................................................................................................... 69
2 Principles of Bioluminescent Enzymatic Bioassays .......................................................... 71
2.1 The Bioassays Based on the Bacterial Coupled Enzyme System:
NADH:FMN-Oxidoreductase-Luciferase................................................................... 71
2.2 Enzymatic Reagents for Bioluminescent Analysis.................................................... 75
2.3 Biotesting Natural and Waste Waters Using the Soluble and Immobilized
Coupled Enzyme System NADH:FMN-Oxidoreductase-Luciferase ........................ 83
2.4 The Reagent ‘‘Enzymolum’’ in Toxicological Bioassay of Air ............................... 85
2.5 Signal System of Enzymatic Assays: Methodological Aspects................................ 86
2.6 How to Design New Bioluminescent Enzymatic Bioassays..................................... 87
2.7 Signal System of Enzymatic Methods for Toxicological Bioassay
of Natural and Laboratory Aquatic Ecosystems ....................................................... 91
2.8 Signal System of Enzymatic Assays for Toxicological Bioassay of Pesticides ...... 99
3 Summary ............................................................................................................................ 103
References................................................................................................................................ 105
Application of Enzyme Bioluminescence 69
1 Introduction
Luciferase ðLÞ
ð1Þ
FMN H2 þ RCHO þ O2 ! FMN þ RCOOH þ H2 O þ hm
that cannot solve the pressing problem of how to detect, identify, and measure the
contents of the numerous chemical compounds that differ in their physicochemical
and toxic characteristics. Moreover, BEST was introduced to facilitate and
accelerate the development of cost-competitive enzymatic systems for use in
biosensors for medical, environmental, and industrial applications.
The present work examines the general principles of bioluminescent enzymatic
toxicity bioassays and describes the applications of these methods and their
implementation for commercial biosensors.
Ic
Analyzed sample
200
Iexp
Ic
Analyzed sample
200
Iexp
P
100 200 300 t, s
Tmax
system of luminous bacteria, their substrates, and buffer solution are placed into a
bioluminometer, and after the maximum light emission intensity Ic (control) is
registered, from 5 to 50 lL of the test water sample or toxic substance solution are
added, and the maximum light emission intensity Iexp is registered again (Fig. 2a).
The intensity of the residual luminescence (Iexp/Ic) 100 % and the decay
constant kd are estimated. The decay constant is calculated according to the fol-
lowing formula: kd = [ln(I2/I1)]/Dt, where I1 is the peak of bioluminescence
intensity, I2 is the bioluminescence intensity at the certain moment of time after
reaching the bioluminescence maximum, and Dt is the time (minutes) needed for I1
to reach I2.
When analyzing toxicity of the water samples, the luciferase index (LI) or
toxicity coefficient (TC) are calculated according to the formulas:
LI ¼ Iexp Ic 100 %;
TC ¼ Ic Iexp Ic 100 %:
LI and TC are the residual luminescence and the degree of inhibition of the
bacterial coupled enzyme system NADH:FMN-oxidoreductase-luciferase in the
presence of analyzed sample correspondingly and TC = 100 - LI.
74 E. Esimbekova et al.
characterizes the spatial and temporal water quality of Lake Shira. A partial map
was based on the bioluminescence characteristics of water samples taken along the
shoreline, sampling stations in the different places, and in different depths of the
lake. The map reflects the correlation between the effect of the water samples on
bioluminescence intensity and their chemical and physical–chemical characteris-
tics. It has been demonstrated that the bioluminescence assay measurements must
be done within 2 h after the sampling time.
The advantages of enzymatic assays based on the bacterial coupled enzyme
system L + R are their rapidity (the time of analysis does not exceed 3–5 min),
high sensitivity, simple measuring procedure, possibility of automation of eco-
logical monitoring procedure, availability and safety of reagents, and a wide
market of bioluminometers [31].
They were so easy to use and convenient that they started to be applied in the
educational process, mainly in ecology practical courses [32–35].
Nylon tube reactor Complex 6–8 50–70 100 (60) G [1 900 Yes
Glass strips Easy 1–2 92–96 100 (60) G1 100 Yes
Cellulose film Easy 1–2 3–8 – G1 – Yes
Lipid films Complex 5–10 0.1–1 Stable (1–7) P— – No
Bovine serum albumin gel Easy \1 0,3–8 100 (30–60) —1 100 No
Insoluble collagen Easy 1–2 100 70 (14) 20 (240)d G— Several Yes
Starch gel Easy 10–12 100 100 (360) VG 0.1 100 Yes
Alginate gel Easy 2–10 30–60 100 (1) G— None No
Polyamide membranes Moderate 2–4 3–10 100 (180) —1 None No
a
Relative activity compared with soluble enzyme
b
Percentage of initial activity after number of days at 4 C
c
Reproducibility: G good; VG very good; P poor, or sensitivity relative to soluble enzyme set 1
d
High mechanical strength
77
78 E. Esimbekova et al.
The following methods were tested to increase the sensitivity of the reagent to
pollutants: varying the composition of the immobilized reagent; varying the ratio
of reaction mixture and test sample; varying the order of components added to the
coupled enzymatic system and test sample; introducing an additional step of
reagent incubation to the test water sample; and selecting the control solution for
biotesting [46].
(a) 100
80
Iexp/Ic,%
60
40
20
0
0.1 0.2 0.4 0.1 0.2 0.4 0.1 0.2 0.4
(b) 80
70
60
50
Iexp/Ic,%
40
30
20
10
0
0.1 0.2 0.4 0.1 0.2 0.4 0.1 0.2 0.4
0.2 µg L 0.5 µg L 1 µg L
[NADH], mM
Fig. 4 Residual light intensity of immobilized reagent under variation of luciferase (L) and
NADH content in reagent in the presence of 50 lg L-1 benzoquinone (a) or 5 lg L-1 CuSO4
(b) [46]
It was shown previously [39] that introducing FMN into the immobilized reagent
resulted in a significant decrease in the activity of the coupled enzymatic system;
that is why FMN was not included in the reagent and FMN solution of various
concentrations was added during the analysis. When the FMN concentration in the
reaction mixture was increased, the sensitivity of the multicomponent reagent to
benzoquinone decreased and remained at the same level to CuSO4 (Fig. 6).
Thus, it is shown that by reducing the content of any component in the
immobilized reagent, it is possible to increase the reagent’s sensitivity to toxic
80 E. Esimbekova et al.
Iexp/Ic,%
was: DTT 0.1 mM, 40
mercaptoethanol 0.2 mM,
BSA 0.001 mM. Control is
the reagent without any
stabilizer [46] 20
0
control DTT mercaptoethanol BSA
20
0
0,008 0,02 0,03 0,08
[FMN], mM
substances. This effect can be explained by the fact that the coupled enzyme
system had nonstationary conditions because the reagent contains a nonsaturating
concentration of one of the substrates (NADH). Indeed, the maximum sensitivity
of the coupled enzyme system to benzoquinone and CuSO4 was observed when the
NADH concentration was 1.3 times lower than the one corresponding to the
Michaelis constant for this substrate [40].
In all the experiments described above, the pollutant solution was added to the
reaction mixture after the maximum luminescence level had been reached, that is,
after the substrates and enzyme active centers had interacted and, probably,
Application of Enzyme Bioluminescence 81
Iexp/Ic,%
benzoquinone: 1 introduction
of analyzed water sample 60
before starting enzyme
reactions by FMN; 2 40
introduction of analyzed
water sample after light
intensity reaches its 20
maximum [46]
0
50 µg/L benzoquinone 5 µg/L CuSO4
The multicomponent reagent with the control or analyzed pollutant solution was
incubated for 30–900 s, then the reaction was started with the FMN solution, and
Ic and Iexp were measured (Fig. 8).
It was shown that such additional incubation increases the sensitivity of the
immobilized reagent to copper sulfate, and the sensitivity to benzoquinone remains
at the same level. The inhibition of enzymes with the copper sulfate solution grew
proportionally to the increase of incubation time and reached its peak when the
reagent had been incubated for 120–130 s with little effect on the assay sensitivity
at 300-s incubation. A further increase of incubation time is impossible because
the activity of the immobilized reagent is significantly lowered, even when the
control solution is analyzed.
Thus, we found the conditions for biotesting aqueous solutions using the
immobilized reagent that provide the maximum sensitivity to toxic substances.
82 E. Esimbekova et al.
Iexp/Ic, %
60
(2) solutions [46]
40
20
2
0
0 50 100 150 200 250 300 350
Time, s
The effect of pollutants upon the bioluminescent system was estimated according
to the parameter EC50 which is the concentration of the active substance when
bioluminescence is inhibited by 50 %. This parameter was chosen by analogy with
the generally accepted parameter EC50 which is the effective concentration of the
active substance causing a 50 % change in some vital functions of the test
organism [47]. The values of EC50 were determined for various classes of organic
pollutants using the soluble and immobilized coupled enzyme system L + R
(Table 2).
Table 2 shows that the sensitivity of the soluble coupled enzyme system L + R
to the studied pollutants was higher as compared to that of the immobilized
Application of Enzyme Bioluminescence 83
efficiency of using the immobilized reagent instead of the soluble coupled enzyme
system L + R in biotests.
Another example is concerned with water bodies containing clean ‘‘reference’’
water, such as Lake Baikal. During the studies of water from Lake Baikal a
comparison was made of assays based on a soluble and immobilized coupled
enzyme system depending on the place and depth of sampling (Fig. 9). Samples
from five control points were studied.
Application of Enzyme Bioluminescence 85
Depth, m
300
400
500
600
The nature of luminous intensity dependence for water samples from different
depths was similar with both soluble and immobilized coupled enzyme systems.
Water samples taken from the upper (the warmest) epilimnion zone didn’t cause
intense luminescence; it must be mentioned that, depending on the location of the
sampling station, the zone was within the range of 5–50 m from the surface.
Apparently, the slight stimulation effect could be explained by the fact that the
epilimnion water, as compared to the other water body layers, contains more
oxygen due to a larger phytoplankton biomass. Water samples from the depth of
50–100 m (depending on the place of sampling) had a slight effect on the luminous
intensity with both soluble and immobilized coupled enzyme systems, which
didn’t suggest pollution, as the test readings were within the range of possible
fluctuations of enzymatic assay parameters for ‘‘normal’’ unpolluted water.
Bioluminescent assays were used not only for aquatic environments, but also for
monitoring of air pollution [49]. The sensitivity of luminous bacteria and their
enzymes to air samples differing by industrial pollution degree was determined.
Air samples were collected in the clean (Akademgorodok, sample #1) and polluted
(the coal power plant area, sample #2) districts of the city of Krasnoyarsk. The air
samples were collected into liquid absorption medium (water, ethanol, or acetone).
The standard aspirating device performing 1.0 L/min was used. Chemical com-
position of the samples was analyzed with a gaseous chromatograph (Agilent
Tech. 7890A). To compare the sensitivity of assays the numbers of dilution of the
samples necessary to remove the toxic effect were considered (Table 6).
The results indicate that water is better than ethanol or acetone medium for air
sample preparation because of its sufficient capacity to absorb organic compounds,
and absence of interfering effects on bioluminescence. The sensitivity of soluble
86 E. Esimbekova et al.
and immobilized enzymes is 3–24 times higher than the sensitivity of bacterial-
based tests. The immobilized reagent provides the reduction of the time required to
complete the analysis (down to 7 min), easy-to-use, higher sensitivity (allowed
dilutions are up to 16,000), and the possibility to increase the volume of the sample
up to 97 % of the total. Thus there is the possibility to apply the bioluminescent
bioassays based on the immobilized reagent Enzymolum for air pollution moni-
toring [49].
In all the existing bioassays based on living organisms, the reaction of the test
substance with the substances that are harmful for these objects is determined. In
this context the term ‘‘environmental toxicity’’ is used, although it is not always
correct. It is also considered that all living organisms, including humans, react with
the test medium in a similar way. Such a conception is based on the statement that
all living organisms have a similar structure and functions that are affected by
toxic substances. The idea that normal vital activity of all living organisms—from
amoebas to humans—is based on complex interconnected activity of many
enzymatic systems providing with such functions as respiration, digestion,
reproduction, bioluminescence, and the like, suggests that a universal test system
could be developed consisting of a number of enzymatic reactions. Indeed, it is
generally accepted that all living beings, despite the diversity of substrates
involved in metabolism, including specific ones for a given organism, have similar
basic types of reactions, and based on the activity of enzymes released under the
effect of pollutants it is possible to determine the biochemical mechanisms of these
substances affecting not only functions, but organisms on the whole.
Application of Enzyme Bioluminescence 87
Such a model enzymatic test system must include enzymes of different classes,
or key enzymes of metabolic processes in living organisms. In this case it will be
possible to determine which vital function of organisms will be inhibited by which
toxic substance or their mixture.
Developing such a complex model test system is possible on the basis of
bioluminescence. Using the coupling principle in bioluminescent analysis gives
almost unlimited possibilities for determining the activity of various enzymes [5]
and designing new enzymatic assays on this basis. The possible basis for devel-
opment of new bioluminescent enzymatic bioassays is the coupled enzyme reac-
tion NADH:FMN-oxidoreductase-luciferase.
The unique feature of the enzymatic analytical systems is that, due to the
coupling with bacterial luciferase, it is possible to measure the activity of more
than 100 enzymes [5] if enzyme coupling chains are created where the product of
one enzyme is the substrate of the next one. The luciferase of luminous bacteria
must be the terminal enzyme in coupling chains for such enzymes as lactic
dehydrogenase, trypsin, glucose-6-phosphate dehydrogenase, and others, making it
possible to measure the activity of many enzymes according to the luminous
intensity.
Moreover, using enzymatic reactions instead of living organisms in toxicity
bioassays makes it possible to regulate the sensitivity of enzymatic assays
depending on the objective. For example, the sensitivity of enzymatic assays to the
toxic substances may be increased by extending the coupling chain of enzymatic
reactions [50].
As a result of using several chains of coupling enzymatic reactions with bac-
terial bioluminescence, a set of enzymatic assays sensitive to different groups of
pollutants was created as a ‘‘signal system of enzymatic assays’’ [25, 51].
(a) FMN
ADH R L
NAD + NADH FMNH 2 Light
(b) Tripsin
FMN
R L
bacteria (Reaction 1). Later a coupled enzymatic bioassay was created by coupling
reaction (1) with NADH:FMN-oxidoreductase (Reaction 2) [11]. In the work of
Kudryasheva et al. [50], three enzymes were coupled with direct ADH reaction as
an example. In this case, NAD+ was added to the reaction mixture that was subject
to enzymatic reduction (Reaction 3) catalyzed by ADH, the resulting product
NADH entered Reaction (2), and then other stages of the light emission process
occurred.
ADH
ð3Þ
C2 H5 OH þ NADþ , NADH þ Hþ þ C2 H4 O
4000 1 kd background
3a
2000 kd exp
2
3b
kd control
kd exp
t, s
50 1000 1500 2000
Fig. 11 Scheme of ADH (or trypsin) activity measurement using the bioluminescence decay
constant: kd background is the decay constant for the coupled enzyme Reaction (1); kd control is the
decay constant for the triple enzyme reaction with ADH or trypsin in the absence of pollutant (2);
kdexp is the decay constant for the triple enzyme reaction with ADH or trypsin in the presence of
pollutant. 3a the pollutant inhibits trypsin or ADH activity; 3b the pollutant activates trypsin or
ADH activity
(a) P, s (b) P, s
40 300
30
200
20
100
10
0 0
0 0,5 1 1,5 2 0 0,05 0,1
C, mM C, µM
Thus, estimation of the effect of chemical substances on the activity of the triple
enzyme system with ADH and trypsin is based on changes in kinetic parameters of
bioluminescence (Fig. 11). The procedure for triple enzyme biotesting is as
follows.
• The maximum luminescence intensity of the coupled enzyme system L + R is
measured, and then its luminescence intensity decrease is measured for 1 min.
The decay constant (kd background) for the case of the coupled enzyme reaction is
calculated by the formula: kd background = ln(Imax/I1)/(t1 - tmax), where Imax is
the bioluminescence intensity maximum and I1 is the bioluminescence intensity
at the moment of time t1 (Fig. 11).
• 5–50-lL test sample and 5-lL ADH or trypsin solution are added to the
reaction mixture described above. Then the decay constant kd exp is measured
for the triple enzyme reaction.
• The authentic decay constant for the triple enzyme reaction is calculated by the
equation: kd = kd exp - kd background.
• For control measurement, 5-lL ADH or trypsin solution and 5–50-lL phos-
phate buffer (0.05 mol L-1, pH 6.8) or distilled water are added to the reaction
mixture described above. The control decay constant, kd control, is calculated.
• Relative activity of ADH or trypsin is calculated using the equation: A = (kd/kd
control) 9 100 %. The obtained value is proportional to the sample’s toxicity.
Fig. 13 Dynamics of the pond ecosystem components: (d) epilimnion, (s) hypolimnion [25]
and hypolimnion temperature. Dissolved oxygen saturation values (Fig. 13) in the
epilimnion of the pond were significantly higher than those in the hypolimnion. In
phytoplankton, green algae Volvox aureus Ehr. and diatom Cyclotella comta (Ehr.)
Kutz. dominated. Blooms of V. aureus took place in the middle of July and in the
middle of August (Fig. 13). C. comta dominated in the middle of June, when the
water temperature was comparatively low. At the end of July and beginning of
August, a small decrease of water temperature took place (Fig. 13) and C. comta
became dominant in this period. In the hypolimnion only two samples of phyto-
plankton were taken in June and July, but the other dominant diatom and green
species then in the epilimnion, Stephanodiscus hantzschii Grun., and Closterium
peracerozum were found in the samples. Thus, the epilimnion and hypolimnion
represented separate compartments of the pond ecosystems and naturally differed
in water quality.
The variations of enzyme bioassay data were insignificant during the study
period (Fig. 14). The data for the luminous bacteria based assay varied over the
season. Water samples taken from the hypolimnion had a stronger inhibitory effect
on bacterial luminescence.
Application of Enzyme Bioluminescence 93
Fig. 14 Parameters of bioluminescent bioassays for the study of water from local pond: (d)
epilimnion, (s) hypolimnion. A, %: relative activity of ADH and trypsin, Ic and Iex: light
intensity in control and experimental cuvette, respectively, for NADH:FMN-oxidoreductase-
luciferase and luminous bacteria bioassays [25]
Only the bioassay based on the luminous bacteria demonstrated the significant
difference between the epilimnion and hypolimnion by the Wilcoxon test. For the
assays based on enzyme systems, the differences between the epilimnion and
hypolimnion were insignificant.
At the end of summer water samples from the pond epilimnion (depth of about
2 ± 3 m) were inoculated into three cylindrical glass experimental microecosys-
tems (MES) 25 cm in diameter and 25 cm high. They were illuminated by white
light of 2.5 W/m2 (natural intensity at the depth of sampling). Water samples from
MES were taken twice a week for 2 months. Benzoquinone was added in the last
week of study to MES 1.
The MES’s experiment consists of two parts. In the first part (lasting 42 days;
Fig. 15a) the relatively higher biomass of microalgae from the pond was
94 E. Esimbekova et al.
Fig. 15 Biomass of algae and the dynamics of bioassay parameters for MES: (d) MES 1
(treatment with benzoquinone on the 35th day of experiment (b), (j) MES 2, (+) MES 3. (a), %:
relative activity of ADH and trypsin, Ic and Iex: light intensity in control and experimental
cuvette, respectively, for bacteria bioassay. a and b are experiments with high and low biomasses
of microalgae, respectively [25]
inoculated into the MES 1–3 and the bioassay reactions at the high biomass were
recorded. In the second part of the experiment (Fig. 15b) MES 1–3 were reinoc-
ulated and the low biomass of microalgae was maintained in MES 1 ± 3.
Application of Enzyme Bioluminescence 95
‘‘Bloom’’ in the water body can also change the response of the bioassays. It was
shown earlier that bloom of blue-green algae was the characteristic feature of the
ecosystem which indicated the distinctive rate and kinetics of the phenol bio-
degradation [60]. No bloom of blue-green algae was observed in the water body
under investigation, thus we used a laboratory culture of blue-green microalgae
Spirulina platensis as a model of a blooming pond.
Dynamics of bioassay data and growth of algae are depicted in Fig. 17. One can
see that throughout the algal growth period, the response of the bioassay based on
the coupled enzyme system varied insignificantly. It is evident from the assay data
96 E. Esimbekova et al.
based on the triple enzyme systems that during the first days of their growth algae
release metabolites inhibiting ADH and enhancing trypsin activity. After 4 days of
algal growth, trypsin activity in the presence of the culture medium did not change.
Response of the bioassays to the algal culture medium was the same in each of the
three replicates. The bioassay based on ADH was the most sensitive. By the eighth
and ninth days, the inhibition of the triple enzyme system with ADH was almost
full (Fig. 17).
Therefore, bioluminescent assays show that their sensitivity differs by water
quality, even in the case of the unpolluted pond. The assay based on luminous
bacteria proved to be the most sensitive. The difference of water quality between
the epilimnion and hypolimnion were due to natural causes rather than by any
pollution. Hence, the result of this assay is determined by the depth at which water
Application of Enzyme Bioluminescence 97
Fig. 17 The growth of microalgae biomass and the bioluminescent bioassay parameters for the
study of the culture medium of S. platensis. (r) effect of culture growth medium at the beginning
of experiment, (s, h, D) three replicates, D680 optical density of S. platensis culture medium at
680 nm (the maximum of chlorophyll), A relative activity of ADH and trypsin, Ic and Iex light
intensity in control and experimental cuvette, respectively, for NADH:FMN-oxidoreductase-
luciferase bioassay [25]
samples were taken (i.e., presence of the water from deoxygenated hypolimnion);
this should be remembered when using this assay for the monitoring of water
bodies.
Changes in the response of bioassays over the period of investigation were not
related to pollution of the water body. Parameters of the bioassays did not change
dramatically even in the periods of bloom of the green algae V. aureus. Indeed, in
98 E. Esimbekova et al.
the unpolluted pond and unpolluted MESs, were significantly lower than the
degree of bioassay response after the addition of the pollutant (benzoquinone).
Thereby we could detect the effect of pollutants such as quinones, within the
variability of responses, caused by natural water.
Sensitivity of the assays to contaminants of polluted or ‘‘blooming’’ pond has
been shown to differ, inasmuch as the mechanisms of xenobiotic (benzoquinone)
action on the assay systems are different. Hence, the data of a single assay cannot
provide a basis for a conclusion about the presence or absence of toxic substances
in a water body. Only a set of assays, such as the one used in the present study can
be applied as an alarm system to detect an acute toxicity of aquatic ecosystems.
Pesticides vary in their toxicity mechanism and character; for example, they can be
carcinogenic or mutagenic, or they can affect the respiratory, endocrine, immune,
or nervous systems [61, 62]. The effect of pesticides (organophosphates and
pyrethroid preparations) on the bioluminescence of the four systems has been
analyzed. The characteristics of the toxic substances analyzed are listed in Table 7.
Four bioluminescence assay systems were used in our studies (Table 8). For
each of the analyzed substances, values of EC50 and EC20 were obtained (Table 8).
They constituted 50 and 20 % of the loss of luminescence for the coupled enzyme
system and luminous bacteria, or 50 and 20 % loss of the relative enzymatic
activity for the triple enzyme systems with ADH and trypsin. The parameter EC20
was taken for the authentic determination of a toxicant’s impact on biolumines-
cence and for the comparison of the toxicant maximum permissible concentration.
The sensitivity of the coupled enzyme system to the impact of organophos-
phorous compounds differed widely. The EC20 of pirimiphos-methyl (Table 8, No.
2), for instance, was 0.9 mg 9 L-1, whereas glyphosate (Table 8, No. 3) had no
effect at all on the bioluminescence of the coupled enzyme system. It should be
noted that glyphosate is a herbicide that has low toxicity to animals. Hence, their
toxicity is in good correlation with bioluminescence inhibition.
The triple enzyme systems with ADH and trypsin are more sensitive to
organophosphorous compounds (Table 8). The sensitivity of luminous bacteria to
the impact of organophosphorous compounds is comparable to that of the coupled
enzyme system (Table 8). For example, the EC20 of pirimiphos-methyl is the same
in both cases, 0.9 mg 9 L-1.
All the studied test systems were very sensitive to pyrethroid substances in the
range 0.1–3 mg 9 L-1 (Table 7, Nos. 4–7). These insecticides, synthetic ana-
logues of natural pyrethrins, act through intestinal contact, thereby affecting the
nervous and the immune systems [63–65]. With pyrethroid, the values of EC20 are
smaller for the bacterial system than for the coupled enzymatic system. To
100 E. Esimbekova et al.
2. Pirimiphos-methyl 305.33 O
3. Glyphosate 169.07 O
4. a-Cypermethrin 416.30 P
5. Bifenthrin 422.87 P
6. Deltamethrin 505.2 P
7. k-Cyhalothrin 449.85 P
compare: with deltamethrin (Table 8, No. 6) the values of EC20 are 0.06 and
0.001 mg 9 L-1 for the coupled enzyme system and luminous bacteria, respec-
tively. This is attributed to permeability and destruction of cell membranes due to
the high lipophilicity of these substances. Sensitivity of the triple enzyme systems
to certain substances is similar to that of luminous bacteria.
Pyrethroid and organophosphorous compounds inhibited trypsin activity.
Organophosphorous compounds are known to have an effect on proteolytic
enzymes [61, 66]. They are able to inhibit esterase and proteolytic activity of both
trypsin and chemotrypsin. This may account for the inhibitory effect of organo-
phosphorous compounds observed in the triple enzyme system with trypsin.
Table 8 The influence of toxic substances on bioluminescent assay systems [51]
Number Substance Luminous bacteria Coupled enzyme system Triple enzyme system with Triple enzyme MPC PDD
photobacterium trypsin system with ADH (mg L-1) (mg kg-1)
phosphoreum
EC50 EC20 EC50 (mg L-1) EC20 EC50 (mg L-1) EC20 EC20 (mg L-1)
Application of Enzyme Bioluminescence
Fig. 18 Relative activity (A) of ADH (1) and trypsin (2) under different concentrations of
deltamethrin. Influence of the pollutant on triple enzyme systems resulted in trypsin inhibition
and ADH activation [51]
3 Summary
cells according to detection of cellular NADH using the coupled enzyme system:
NADH:FMN-oxidoreductase-luciferase.
Integrated bioluminescent methods are also very promising for medical
research, for example, for evaluating the gravity of endotoxicosis during treatment
in surgery and therapy. The methods are highly sensitive, rapid, and simple and
allow quantitative determination of the degree of seriousness of illness and the
estimation of the severity of a patient’s condition, disease course prediction,
estimation of the efficiency of the used detoxification methods, and determination
of the optimal operation mode of drainage facilities made of semipermeable
membranes [82–84]. Controlled perfusion of rats’ liver demonstrated a possibility
of using an integrated bioluminescent method for the intensity assessment of
pathologic oxidation processes [85]. A very interesting and promising trend in the
development of integrated bioluminescent testing is the creation of rapid analysis
for the assessment of human organism reaction to physical and mental stress.
Analysis is made by comparing the light emission intensity of the coupled enzyme
system NADH:FMN-oxidoreductase-luciferase in the presence of a person’s saliva
taken before and after a certain stress load [86]. The main advantages of the
bioluminescent method are not only its rapidity, high precision, and sensitivity, but
also noninvasiveness, because human saliva is analyzed, which reflects the func-
tional state of a person just as blood does.
Thus, the new approach of biotechnological design developed on biolumines-
cent enzymatic biosensors, methods, and reagents has been described in the
chapter. To solve the problem of how to detect, identify, and measure the contents
of the numerous chemical compounds in environmental monitoring, food product
monitoring, and medical diagnostics, we proposed Bioluminescent Enzyme Sys-
tem Technology BESTTM, wherein the bacterial coupled enzyme system:
NADH:FMN-oxidoreductase-luciferase substitutes for living organisms. BESTTM
was introduced to facilitate and accelerate the development of cost-competitive
enzymatic systems for use in biosensors.
A patented stabilization and immobilization process produces the multicom-
ponent reagent Enzymolum, which contains the bacterial luciferase, NADH:FMN-
oxidoreductase, and their substrates, coimmobilized in starch or gelatin gel. The
reagent is currently produced in tablet form and can be used only in the cuvette
variant of a bioluminometer. The other forms, for example, on the plane table,
strips, and others were also introduced for bioluminescent analysis. Enzymolum
can be integrated as a biological module into the portable biodetector–biosensor of
original construction.
Enzymolum is the central part of the Portable Laboratory for Toxicity Detection
(PLTD), which consists of a biological module, a biodetector module, a sampling
module, a sample preparation module, and a reagent module. PLTD allows us (a)
to detect a wide range of toxic substances, more than 25,000 compounds; (b) to
perform rapid screening for toxicity in emergency situations in the field and lab-
oratories; (c) to develop systems for analyzing individual compounds; (d) to
develop systems to evaluate the degree of overall toxicity; (e) to keep the high
sensitivity of reagents for many years; and (f) to perform biotesting in the presence
Application of Enzyme Bioluminescence 105
of high concentrations of organic substances in water. The last item suggests the
uniqueness of enzymatic bioassays, because no bioassays based on living organ-
isms give consistent results under such conditions. Living organisms use organic
substances for food and thus bioassays based on living organisms do not reflect
environmental quality correctly, resulting in the absence of accuracy and reliability
of analysis.
Prototype biosensors developed with this technology offer cost advantages,
versatility, high sensitivity (up to 10-14 mol of analyte), rapid response time (less
than 3 min), extended shelf life (up to 2 year), and flexible storage conditions (up
to +25 C).
The enzyme biotesting approach was used as a platform technology to certify
‘‘Method to measure the intensity of bioluminescence with the help of the ‘Enz-
ymolum’ reagent to detect the toxicity of drinking, natural, waste, and treated
waste water’’ [48]. The laboratory will be the principal example of a whole family
of new, portable, professional laboratories for ecological monitoring, food quality
laboratories, military departments, and other monitoring, teaching, security, and
research organizations.
Acknowledgments The work was financially supported by the Government of the Russian
Federation (Contract No 11. G34.31.0058) and Project 1762 from the Ministry of Education and
Science of Russian Federation.
References
12. Gil TA, Belesova NP, Balayan AE et al (1988) Determination of the activity of
phenoloxidases in solution. RU Patent 1,557,521, 1 Dec 1988
13. Kratasyuk VA, Kruchinina RI, Kuznetsov AM et al (1985) The method to determine
concentration of the inhibitors of biological activity. RU Patent 1,204,639, 5 Sept 1985
14. Kratasyuk VA, Kruchinina RI, Kuznetsov AM et al (1986) The method to determine
concentration of acrylonitrile. RU Patent 1,270,658, 15 Jul 1986
15. Kratasyuk VA, Kuznetsov AM, Fish AM et al (1981) The method to determine concentration
of the inhibitors of biological activity. RU Patent 865,904, 23 Sept 1981
16. Kudryasheva NS, Kratasyuk VA, Esimbekova EN et al (1998) Development of the
bioluminescent bioindicators for analyses of environmental pollutions. Field Anal Chem
Tech 2:277–280
17. Kudryasheva N, Vetrova E, Kuznetsov A et al (2002) Bioluminescent assays: effects of
quinones and phenols. Ecotox Environ Safe 53:221–225
18. Vetrova EV, Kudryasheva NS, Kratasyuk VA (2007) Redox compounds influence on the
NAD(P)H:FMN-oxidoreductase-luciferase bioluminescent system. Photoch Photobiol Sci
6:35–40
19. Kudryasheva NS (2006) Bioluminescence and exogenous compounds. Physico-chemical
basis for bioluminescent assay. J Photochem Photobiol, B 83:77–86
20. Kudryasheva NS (2006) Nonspecific effects of exogenous compounds on bacterial
bioluminescent enzymes: fluorescence study. Curr Enzym Inhib 2:363–372
21. Kratasyuk VA, Fish AM (1980) Study of mechanism of 2,4-dinitrofluorobenzene effect on
bacterial luminescence in vitro. Biochemistry (Moscow) 45:1175–1181
22. Kratasyuk VA, Makurina VI, Kuznetsov AM et al (1991) Study of effect of sulfo-substituted
succinic acid on bacterial luminescence. Appl Biochem Micro (Moscow) 27:127–133
23. Kudryasheva NS, Esimbekova EN, Yu Kudinova I et al (2000) Effects of quinones on
NADH-dependent enzymatic bioluminescent systems. Appl Biochem Micro (Moscow)
36:409–413
24. Kratasyuk VA, Esimbekova EN (2011) Express method for biotesting of natural,
manufacturing waters and water solutions, RU Patent 2,413,771, 10 Mar 2011
25. Kratasyuk VA, Esimbekova EN, Gladyshev MI et al (2001) The use of bioluminescent
biotests for study of natural and laboratory aquatic ecosystems. Chemosphere 42:909–915
26. Kratasyuk VA, Kuznetsov AM, Rodicheva EK et al (1996) Problems and prospects of
bioluminescence assays in ecological monitoring. Sib J Ecol 5:397–403
27. Kratasyuk VA, Vetrova EV, Kudryasheva NS (1999) Bioluminescent water quality
monitoring of salt Lake Shira. Luminescence 14:193–195
28. Kudryasheva N, Shilova E, Khendogina E et al (1999) Lake Shira, a Siberian salt lake:
ecosystem, structure and functions. 3: The use of bioluminescent biotests to monitor
ecological status. Int J Salt Lake Res 8:245–251
29. Vetrova EV, Kratasyuk VA, Kudryasheva NS (2002) Bioluminescent characteristics of Shira
Lake water. Aquat Ecol 36:309–315
30. PD 53.18.24.83-89 (1990) Methods of estimation of kinetic indices of surface water quality.
Gidrometeoizdat, Moscow
31. Deryabin DG (2009) Bacterial bioluminescence: base and applied aspects. Science, Moscow
32. Gitelson JI, Kratasyuk VA (2002) Bioluminescence as an educational tool. In: Kricka LJ,
Stanley PE (eds) Bioluminescence and chemiluminescence: progress and current
applications. World scientific publishing, River Edge, pp 175–182
33. Kratasyuk VA, Gusev SM, Remmel NN et al (2007) Bioluminescence in the spaceflight and
life science training program at Kennedy space center. In: Szalay A, Hill P, Kricka L, Stanley
P (eds) Bioluminescence and chemiluminescence: chemistry, biology and applications.
World scientific publishing, San Diego, pp 257–260
34. Kratasyuk VA, Kuznetsov AM, Gitelson JI (1997) Bacterial bioluminescence in ecological
education. In: Hastings JW, Kricka ZJ, Stanley PE (eds) Bioluminescence and
chemiluminescence (molecular reporting with photons). Willey, Chichester, pp 177–180
Application of Enzyme Bioluminescence 107
57. Stom D (1977) Influence of polyphenols and quinones on aquatic plants and their blocking of
SH-groups. Acta Hydrochim Hydrobiol 5:291–298
58. Kudryasheva NS, Kratasyuk VA, Belobrov PI (1994) Bioluminescent analysis. The action of
toxicants: physical-chemical regularities of the toxicants effects. Anal Lett 27:2931–2947
59. Kudryasheva N, Shalaeva E, Zadorozhnaya E et al (1994) Patterns of inhibition of bacterial
bioluminescence in vitro by quinones and phenols components of sewage. Biofizika
39:441–451
60. Gladyshev M, Sushchik N, Kalachova G et al (1998) The effect of algal blooms on the
disappearance of phenol in a small forest pond. Water Res 32:2769–2775
61. Galloway T, Handy R (2003) Immunotoxicity of organophosphorous pesticides.
Ecotoxicology 12:345–363
62. Hansen OCh (2004) Quantitative structure–activity relationships (QSAR) and pesticides.
From Danish ministry of the environment: environmental protection agency. Pesticides Res
94:134. http://www2.mst.dk/udgiv/publications/2004/87-7614-434-8/pdf/87-7614-435-6.pdf
63. Coles GC, Stafford KA (1999) The in vitro response of sheep scab mites to pyrethroid
insecticides. Vet Parasitol 83:327–330
64. Diel F, Horr B, Borck H et al (2003) Pyrethroid insecticides influence the signal transduction
in T helper lymphocytes from atopic and nonatopic subjects. Inflamm Res 52:154–163
65. Pauluhn J, Machemer LH (1998) Assessment of pyrethroid-induced paraesthesias:
comparison of animal model and human data. Toxicol Lett 97:361–368
66. Lundebye AK, Curtis TM, Braven J et al (1997) Effect of the organophosphorous pesticide,
dimethoate, on cardic and acetylcholinesterase (AChE) activity in the shore crab Carcinus
menaeus. Aquat Toxicol 40:23–36
67. Hernando MD, Ejerhoon M, Fernandez-Alba AR et al (2003) Combined toxicity effects of
MTBE and pesticides measured with Vibrio fishery and Daphnia magna bioassays. Water Res
37:4091–4098
68. Rahman MF, Mahboob M, Danadevi K et al (2002) Assessment of genotoxic effects of
chloropyriphos and acephate by the comet assay in mice leucocytes. Mutat Res 516:139–147
69. Datta M, Kaviraj A (2003) Acute toxicity of the synthetic pyrethroid deltamethrin to
freshwater catfish Clarias gariepinus. Bull Environ Contam Toxicol 70:296–299
70. Grisolia CK (2002) A comparison between mouse and fish micronucleus test using
cyclophosphamide, mitomycin C and various pesticides. Mutat Res 518:145–150
71. Strachan G, Preston S, Maciel H et al (2001) Use of bacterial biosensors to interpret the
toxicity and mixture toxicity of herbicides in freshwater. Water Res 35:3490–3495
72. Trajkovska S, Tosheska K, Aaron JJ et al (2005) Bioluminescence determination of enzyme
activity of firefly luciferase in the presence of pesticides. Luminescence 20:192–196
73. Kudryasheva NS, Kratasyuk VA, Rozhko TV et al (2007) Bioluminescent method for
monitoring the radiotoxicity of solutions. Patent WO 2008,036,000, 14 May 2007
74. Rozhko TV, Kudryasheva NS, Kuznetsov AM et al (2007) Effect of low-level a-radiation on
bioluminescent assay systems of various complexity. Photochem Photobiol Sci 6:67–70
75. Fedorova ES, Kudryasheva NS, Kuznetsov AM et al (2007) Bioluminescent monitoring of
detoxication processes: activity of humic substances in quinone solutions. J Photochem
Photobiol, B 88:131–136
76. Kudryasheva NS, Fedorova ES (2009) Bioluminescent assay to determine antioxidant
activity of humic substances. RU Patent 2,376,380, 20 Dec 2009
77. Girotti S, Muratori M, Fini F et al (2000) Luminescent enzymatic flow sensor for D- and L-
lactate assay in beer. Eur Food Res Technol 210:216–219
78. Kratasyuk VA, Egorova OI, Esimbekova EN et al (1998) A biological luciferase test for the
bioluminescent assay of wheat grain infection with Fusarium. Appl Biochem Micro
(Moscow) 34:622–624
79. Kratasyuk VA, L’vova LS, Egorova OI et al (1998) Effect of Fusarium mycotoxins an
bacterial bioluminescence system in vitro. Appl Biochem Micro (Moscow) 34:190–192
80. Kratasyuk VA, Plotnikova NB, L’vova LS et al (1989) Micro fungi bioluminescent assay of
grain infection. RU Patent 1,469,866, 15 Dec 1989
Application of Enzyme Bioluminescence 109
81. Mei C, Wang J, Lin H et al (2009) Quantitative detection of NADH by in vitro bacterial
luciferase bioluminescent. Wei Sheng Wu Xue Bao 49:1223–1228
82. Esimbekova EN, Kratasyuk VA, Abakumova VV (1999) Bioluminescent method non-
specific endotoxicosis in therapy. Luminescence 14:197–198
83. Kratasyuk VA, Kovalevskiy AN, Voevodina TV et al (1991) Determination of patients’ state
under endotoxicosis of infectious genesis. USSR Patent 1,663,548, 15 Mar 1991
84. Sovtsov SA, Kratasyuk VA (1991) Determination of endotoxicosis under surgery. USSR
Patent 1,714,512, 22 Oct 1991
85. Remmel NN, Kratasyuk VA, Maznyak OM et al (2003) Bioluminescent analysis of intensity
of pathological oxidative processes in cells of perfused rat liver after hyperthermia. B Exp
Biol Med (Moscow) 135:43–45
86. Gritsenko EV, Borodulin SV, Bytev VO et al (1996) Bioluminescent control of training
process. In: Materials of the 7th All-Russian conference on homeostasis, Krasnoyarsk, 17–22
March 1996
Detection of Organic Compounds
with Whole-Cell Bioluminescent Bioassays
Tingting Xu, Dan Close, Abby Smartt, Steven Ripp and Gary Sayler
Abstract Natural and manmade organic chemicals are widely deposited across a
diverse range of ecosystems including air, surface water, groundwater, wastewater,
soil, sediment, and marine environments. Some organic compounds, despite their
industrial values, are toxic to living organisms and pose significant health risks to
humans and wildlife. Detection and monitoring of these organic pollutants in
environmental matrices therefore is of great interest and need for remediation and
health risk assessment. Although these detections have traditionally been per-
formed using analytical chemical approaches that offer highly sensitive and spe-
cific identification of target compounds, these methods require specialized
equipment and trained operators, and fail to describe potential bioavailable effects
on living organisms. Alternatively, the integration of bioluminescent systems into
whole-cell bioreporters presents a new capacity for organic compound detection.
These bioreporters are constructed by incorporating reporter genes into catabolic
or signaling pathways that are present within living cells and emit a biolumines-
cent signal that can be detected upon exposure to target chemicals. Although
relatively less specific compared to analytical methods, bioluminescent bioassays
are more cost-effective, more rapid, can be scaled to higher throughput, and can be
T. Xu G. Sayler
Joint Institute for Biological Sciences, The University of Tennessee,
Knoxville, TN, USA
D. Close
Biosciences Division, Oak Ridge National Laboratory,
Oak Ridge, TN, USA
A. Smartt S. Ripp G. Sayler
Center for Environmental Biotechnology, The University of Tennessee,
Knoxville, TN, USA
A. Smartt S. Ripp G. Sayler (&)
Department of Microbiology, The University of Tennessee, Knoxville, TN, USA
e-mail: sayler@utk.edu
designed to report not only the presence but also the bioavailability of target
substances. This chapter reviews available bacterial and eukaryotic whole-cell
bioreporters for sensing organic pollutants and their applications in a variety of
sample matrices.
Contents
1 Introduction........................................................................................................................ 113
2 Detection of Organic Compounds Using Bacterial Bioluminescent Assays .................. 114
2.1 Benzene, Toluene, Ethylbenzene, and Xylene ........................................................ 115
2.2 Polycyclic Aromatic Hydrocarbons ......................................................................... 120
2.3 Alkane Aliphatic Hydrocarbons ............................................................................... 122
2.4 Chlorinated Aliphatic Hydrocarbons........................................................................ 124
2.5 Biphenyl and Polychlorinated Biphenyls................................................................. 125
2.6 Phenol and Derivatives............................................................................................. 127
3 Evaluation of Organic Toxicant-Induced Health Risks Using Eukaryotic Cell-Based
Bioluminescent Assays...................................................................................................... 129
3.1 Bioassays for Dioxin and Dioxin-Like Compounds ............................................... 130
3.2 Bioassays for Hormonally Active Chemicals.......................................................... 135
3.3 Environmental Applications ..................................................................................... 139
4 Conclusions........................................................................................................................ 141
References................................................................................................................................ 142
1 Introduction
Being genetically easy to manipulate and displaying rapid and robust growth,
bacteria have been extensively employed as hosts for bioreporter development.
Despite the toxic nature of organic pollutants, evolution has provided some bac-
teria with unique genetic properties that allow them to adapt to the presence of
toxic chemicals by utilizing them as carbon sources. As the generation of proteins
involved in the degradation of exposed pollutants is an energy-consuming process
and costs fitness under unexposed conditions, the catabolic process is carefully
regulated at the transcriptional level in such a way that it is only initiated upon
exposure to its corresponding chemical input. To exploit this unique feature,
bacterial bioluminescent bioreporters are constructed by transcriptionally
integrating reporter genes (luxAB, luxCDABE, or luc) within special catabolic
pathways that specifically respond to the presence of target compounds. Fortu-
nately, the diversity of catabolic capabilities and sophisticated characterization of
responsible genetic components has provided a sizable toolbox for bioreporter
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 115
Reporter strain Reporter Target compound Detection limit References Environmental application
construct
E. coli DH5a (pPROBE- tubT-luxAB BTEX 0.24 lM toluene [14] Simulated aquatic oil spill [14]
LuxAB-TubT)
E. coli DH5a pGLTUR xylR-Pu-luc Benzene, Toluene, 10–20 lM toluene [13] Soil [13, 131]
Xylene 3 lM 3-xylene
P. putida TVA8 tod-luxCDABE BTEX 0.03–50 ppm BTEX [15] Soil [3]
TCE 1 lM TCE Water [18]
E. coli HMS174 (pOS25) ipbR-luxCDABE Hydrophobic 1 lM toluene [22] Soil [132]
compounds 0.1 lM naphthalene
E. coli DH5a pTOLLUX pR/xlyR-Pu- BTEX N/A [11] Soil and groundwater [11]
luxCDABE
B. sartisoli RP007 (pPROBE- phnS-luxAB PAH 0.17 lM naphthalene [14] Simulated aquatic oil spill [14]
phn-luxAB)
P. putida pPG7-JAMA21 nahR-Psal-luxAB Naphthalene 0.50 lM naphthalene vapor [133] N/Aa
P. fluorescens HK44 nahR-PnahG- PAH 12–120 lM [29] Soil [32, 131, 134]
(pUTK21) luxCDABE
E. coli DH5 (pGEc74, alkS-PalkB-luxAB 6–10-carbon 25 nM octane [37] Groundwater [20]
pJAMA7) alkane Soil [131]
E. coli alkBFG, alkJ- 5–12-carbon 10 lM octane [39] N/A
luxAB alkane
Acinetobacter baylyi ADP1 alkR-PalkM- Alkane 3 nM octane [43] Seawater [43]
luxCDABE
E. coli alkR-PalkM- Alkane 5 nM octane [45] N/A
luxAB
P. putida F1G4 sep-luxCDABE TCE 1 mM TCE [50] N/A
Methylobacterium extorquens dcmR-PdcmA- DCM 1 lM DCM [51] N/A
luxCDABE
(continued)
T. Xu et al.
Table 1 (continued)
Reporter strain Reporter Target compound Detection limit References Environmental application
construct
Ralstonia eutropha ENV307 bphR1- PCBs 0.80 lM 4-chlorobiphenyl [53] N/A
(pUTK60) luxCDABE 4.6 lM Aroclor
E. coli XL1-Blue PpcbC-luxCDABE PCBs 0.1 mM biphenyl (lowest [56] N/A
concentration tested)
E. coli XL1-Blue PpcbC-luc PCBs 0.1 mM biphenyl (lowest [56] N/A
concentration tested)
E. coli (pHYBP109) hbpR-PhbpC- OH-PCBs 1 nM 2-hydroxy-30 ,40 ,5- [58] Simulated aquatic oil spill [14]
luxAB trichlorobiphenyl Human serum [58]
P. fluorescens dmpR-Po- Phenolic 0.30 lM 2-methylphenol [63] Groundwater and semicoke-dump
OS8(pDNdmpRlux) luxCDABE compound 0.87 lM phenol leachates [63]
E. coli (pRLuc42R) luc Phenolic 0.5 lM phenol [65] N/A
compound
Acinetobacter DF4-8 mopR- Phenolic 0.03 mM phenol [67] N/A
luxCDABE compound
a
N/A not available
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays
117
118 T. Xu et al.
bioluminescence was detected above the minimal detection limit after 3 h, and
continued to produce a response after 5 days, demonstrating a longevity of signal
that can be crucial for environmental monitoring applications.
complex nature of diesel oil, a coassay was required to determine the level of
inhibition caused by nonalkane chemicals, which then allowed a corrected octane
equivalency to be determined. Although this assay format was not ideal, it rep-
resented the first time that a bioluminescent microbial assay was used to monitor
for environmental alkane contamination, and provided a valuable first step toward
the development of improved sensor moieties.
Building upon this system, Minak-Bernero et al. [39] were able to take further
advantage of the remaining P. oleovorans alk operon genes and develop an alkane
sensor that did not require the exogenous addition of decanal in order to produce a
bioluminescent signal. To accomplish this, they constitutively co-expressed the
alkBFG alkane catabolism genes, the alkJ alcohol dehydrogenase gene, and the
luxAB genes. Under this system, the alkBFG gene products performed their native
function of reducing the target alkanes to alcohols, the alkJ gene product then
converted those alcohols into aldehydes, and the LuxAB luciferase proteins then
used the resultant aldehydes as substrates for the generation of a bioluminescent
signal. The ultimate result was a bioluminescent microbial bioreporter that could
respond to the presence of alkanes within seconds after exposure to produce a
detectable signal. This sensor was approximately as sensitive as the decanal-
dependent sensor developed previously [37], giving a linear response to octane in
the 10- to 200-lM range [39]. Additionally, because the P. oleovorans alk gene
products were capable of modifying 5- to 12-carbon chain length primary alcohols
and aldehydes [40], and the LuxAB luciferase protein could accept 6-carbon and
longer chain length aldehydes [41], this sensor was theoretically capable of
sensing any alkane between pentane and dodecane. However, because the system
constitutively expressed all of the alk and lux genes, the sensor would report the
detection of any bioavailable pathway intermediate products indiscriminately. This
made it impossible to differentiate alkanes, alcohols, or aldehydes in a given
sample. So although detection had become increasingly autonomous, the
specificity of the system was reduced.
In 2010 there was a renewed interest in the detection of alkane hydrocarbons in
seawater due to the highly publicized Deepwater Horizon oil spill in the Gulf of
Mexico [42]. This spurred renewed testing with the available reporter strains and
demonstrated that, although the detection characteristics were similar to those
obtained in groundwater samples [37], the reporters could be inhibited by salt
during in situ analysis with laboratory-contaminated seawater [14], limiting their
use under environmental conditions. To overcome this deficiency, Zhang et al.
[43] developed a bioluminescent reporter using the native alk operon system in
Acinetobacter baylyi ADP1 rather than expressing a modified version of the
P. oleovorans alk operon in E. coli as had been done previously. This reporter was
constructed using homologous recombination to introduce the luxCDABE genes
from Photorhabdus luminescens in place of the alkM alkane hydroxylase gene in
the A. baylyi ADP1 chromosome, placing them under the control of the naturally
alkane-inducible AlkR regulator protein. Although the detection limit was higher
than that of P. oleovorans-based E. coli reporter strains [37], the A. baylyi ADP1
reporter strain was capable of detecting alkanes between 7- and 36-carbons in
124 T. Xu et al.
More recently, a second P. putida operon has been discovered that can be used
as an alternative to the traditional tod-based approach. This operon, the sep operon,
consists of three efflux pump-encoding genes that are regulated in response to a
variety of common chemical solvents [50]. When the luxCDABE operon was
cloned in place of the upstream sepA gene, the result was a strain that modulated
bioluminescent activity in response to TCE availability. This strain was still
susceptible to interference by the same contaminant chemicals as the TVA8 strain
[15], however, it did present investigators with another tool for the realtime
detection and monitoring of a wide range of halogenated solvents and chlorinated
aliphatic hydrocarbons.
Since the time the tod and sep-based reporter systems were first developed, a
more specific reporter has emerged that can sense and respond to the presence of
the chlorinated aliphatic hydrocarbon dichloromethane (DCM). The selectivity of
this reporter is due to its utilization of the dcm operon from Methylobacterium
extorquens DM4, which is able to grow on DCM as a sole carbon source. The dcm
operon consists of the genes dcmAR, with the dcmA gene upregulated in the
presence of DCM and the dcmR gene encoding a contrasting negative regulatory
element. By cloning the luxCDABE genes under control of the dcm promoter, it
was possible to elicit a bioluminescent response from aerosolized DCM at a range
between 12 lM and 1.2 mM. Induction of the bioluminescent signal could be
observed at 1 h posttreatment at the 1.2-mM level, but increased to 2.3 h at the
12-lM level. In the liquid phase, the reporter could detect DCM between a range
of 1.2 lM–12 mM, and the induction time was relatively decreased compared to
aerosolized samples, requiring only 0.5 h at the 12-mM concentration. Regardless
of the medium used (aerosol or liquid) there was a correlation between biolumi-
nescent output and DCM concentration at an R2 value of 0.99 [51]. In contrast to
the nonspecific reaction of tod- and sep-based systems, this level of specificity and
dose–response kinetics highlights what can be achieved by modulating the
selectivity of the upstream regulatory element that is used for bioreporter
generation.
PCBs represent some of the most widely distributed and persistent environmental
contaminants due to their resistance to physical, chemical, and biological degra-
dation. It is because of this exceptional stability that PCBs found extensive use as
dielectric and coolant fluids in transformers, capacitors, and electric motors.
Evidence of their consequent discharge can be found in nearly all environmental
ecosystems, including water, sediments, soils, and air, and their tendency to bio-
accumulate in living organisms magnifies their presence throughout the food chain
as well. Microbiologically, there are a handful of known bacterial species that can
utilize biphenyl as a sole source of carbon and energy, predominantly via oxidative
degradation mediated by the biphenyl gene cluster (bph) [52]. Layton et al. [53]
126 T. Xu et al.
were the first to exploit the bph pathway for PCB bioluminescent biosensing by
linking the bph R1 regulatory region to a plasmid-localized luxCDABE gene
cassette that was inserted into Ralstonia eutropha to create the bioreporter
ENV307(pUTK60). Validation was performed against biphenyl, 2-, 3-, and
4-chlorobiphenyl, and an Aroclor PCB mixture. However, due to the poor aqueous
solubility of PCBs, a surfactant was added to the test samples to promote increased
bioavailability. Minimum detection limits ranged from 0.80 lM for 4-chlorobi-
phenyl to 4.6 lM for Aroclor in a 96-well microtiter plate assay over a 6-h
incubation period. The need to add surfactants to PCB samples then drove this
group to create an improved toxicity-based bioassay because, in the standard
Microtox test, the toxicity of the surfactants toward the A. fischeri reporters
interferes with the toxicity profile of the PCB compounds [54]. To circumvent this
limitation, Layton et al. [55] developed their toxicity assay using indigenous
wastewater microorganisms displaying surfactant resistance (Stenotrophomonas
sp. and Alcaligenes eutrophus) that were engineered to bioluminesce constitutively
via plasmid insertion of a luxCDABE gene cassette. Results showed these two
strains to be 400 times more resistant than A. fischeri to the commonly used
surfactant polyoxyethylene 10 lauryl ether, signifying their potential practicality in
PCB and other compound toxicity bioassays that require the addition of
surfactants.
Pseudomonas sp. DJ-12 expresses a meta-cleavage dioxygenase via the
pcbABCD operon that enables degradation of select biphenyl compounds. Park
et al. [56] created plasmid-based gene fusions between the pcbC promoter and
luxCDABE and luc to create two bioluminescent bioreporters in E. coli host cells.
Bioassays performed in 96-well microtiter plates over 30-min exposure periods
indicated responsiveness to biphenyl compounds between the 0.1 and 1-mM
exposure concentrations analyzed. Testing at lower concentrations to establish true
detection limits still needs to be performed, as well as compound specificity
studies, but these bioreporters demonstrate a potential addition to the inventory of
biphenyl-responsive bioreporters.
In bacteria and higher organisms, PCBs are biotransformed by cytochrome
P-450 monooxygenases and metabolized to hydroxylated PCBs (OH-PCBs). Cer-
tain bacteria are able to use hydroxybiphenyls as sole carbon and energy sources
via mediation of the hbp gene cluster under regulatory control of the hbpR gene
[57]. Recognizing this, Turner et al. [58] linked the hbpR gene from Pseudomonas
azelaica to the luxAB genes on a plasmid-based (pHYBP109) system that was
inserted into E. coli to create a bioluminescent reporter for OH-PCBs. The biore-
porter was tested against 27 OH-PCBs with dose-dependent responses successfully
obtained with limits of detection in the range from 10-5 to 10-9 M in 4-h incu-
bation assays followed by the addition of the n-decanal substrate. The bioreporter
was also used by Tecon et al. [14] in a luxAB-based multibioreporter assay to
monitor for oil spill constituents in aquatic ecosystems, subsequently allowing for
the simultaneous detection of biphenyls, short-chain linear alkanes, and
monoaromatic and polyaromatic compounds within a 3-h assay. Validation of the
bioreporter was also applied diagnostically in human serum samples spiked with
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 127
Phenols and their derivatives serve as some of the most common environmental
pollutants in soil, water, and air. Deposition occurs through both natural events
(i.e., decomposition of organic material, forest fires, and atmospheric degradation
of benzene) and industrial activities where it is produced in massively high vol-
umes (approximately 7 billion kg per year) as an important precursor component
of plastics, epoxies, explosives, detergents, herbicides, and pharmaceutical drugs.
Under such large-scale manufacturing demands, environmental impacts
(especially in relation to wastewater discharges) are well recognized, with 12
phenolic compounds registered on the US Environmental Protection Agency’s list
of priority pollutants [phenol, 2-chlorophenol, 2,4-dichlorophenol, 2,4-dimethyl-
phenol, 2,4-dinitrophenol, 4-nitrophenol, 2-nitrophenol, pentachlorophenol,
2,3,4,6-tetrachlorophenol, 2,4,6-trichlorophenol, 4-chloro-3-methylphenol (syno-
nym 4-chlor-m-cresol), and 2-methyl-4,6-dinitrophenol (synonym 4,6-dinitro-o-
cresol)]. Accordingly, microorganisms have evolved to utilize these phenols,
which provides an inroad for the development of bioreporter assays based on their
genetic pathways to detect and monitor these phenolic compounds [60]. Over two
decades ago, Shingler et al. [61] isolated a Pseudomonas strain, CF600, capable of
using specific phenols and derivatives as sole sources of carbon. Elucidation of its
genetic pathway for doing so described the now well-understood dmp operon [62]
that later became the platform for the luxCDABE-based bioluminescent bioreporter
P. fluorescens OS8(pDNdmpRlux) [63]. Reporter OS8(pDNdmpRlux) demon-
strated a response portfolio to a variety of phenols, including 2-, 3-, and 4-
methylphenol (synonyms o-, m-, and p-cresol), 2,3-, 2,4-, 3,4-, and 2,6-dimeth-
ylphenol, resorcinol, and 5-methylresorcinol, with maximum detection limits
achieved under 2-methylphenol (0.30 lM) and phenol (0.87 lM) exposures of a 4-
h duration. As is customary with these types of bioreporters, specific phenols
cannot be individually identified and the bioluminescent signal rather represents
the total phenolic content in its bioavailable form. When applied to natural, mixed
contaminant groundwater and semicoke dump leachates containing primarily
128 T. Xu et al.
Table 2 Eukaryotic cell-based bioluminescent bioreporters for dioxin-like chemicals and recent
environmental applications
Host cell Reporter construct Detection EC50 References Recent
(organism) (reporter name) limit (TCDD) (TCDD) environmental
application
S. cerevisiae hAhR and hARNT, 1 nM 4.8 nM [91] Sewage sludge
DRE(95)-luc [135]
Sediment [91]
HepG2 DRE(94)-luc 1 pM 0.35 nM [82] Solid municipal
(human) waste [136]
H4IIE (rat) DRE(94)-luc 0.5 fM 10 pM [83] River water [137]
(H4IIE.Luc) Sediment [137]
Hepa1 DRE(94)-luc 0.1–1 pM 30 pM [80] Storm water [138]
(mouse) (H1L1.1c2) Seawater [92]
RTH-149 DRE(94)-luc 4 pM 64 pM [139] Seawater [140]
(rainbow (RTL 2.0)
trout)
RHEK-1 DRE(94)-luc 10 pM 200 pM [141] N/Aa
(human) (HKY1.7)
Hepa1 DRE(920)-luc 0.01 pM 10–16 pM [142] N/A
(mouse) (H1L7.5c3)
a
N/A not available
In their most basic form, dioxins are any compound containing a heterocyclic
6-membered ring consisting of 2 oxygen atoms and 4 alternative atoms. Practi-
cally, dioxins are persistent pollutants that can bioaccumulate over time, leading to
increased health risks for organisms of higher trophic levels, with the classic
example being 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). TCDD is the most
toxic of the known dioxins, and is thus used as a representative model, with the
relative toxicity of other chemicals expressed in toxic equivalency factors [77].
Collectively, chemicals that inflict toxic effects similar to TCDD are classified as
dioxin-like, and can lead to hepatotoxicity, embryotoxicity, teratogenicity,
immunotoxicity, dermal toxicity, carcinogenesis, or lethality [78, 79]. Dioxin-like
activities have been found in various groups of organic compounds, including
polychlorinated dibenzo-p-dioxins (PCDDs), polychlorinated dibenzofurans
(PCDFs), some PCBs, and PAHs.
The mechanism of action for dioxin-regulated gene expression begins when the
chemical binds to the aryl hydrocarbon receptor (AhR) in the cytosol. This
chemical–AhR complex then translocates into the nucleus and forms a dimer with
an AhR nuclear translocator (ARNT) protein. The dioxin:AhR:ARNT complex
then binds to specific DNA sequences called dioxin-responsive elements (DREs),
which results in the activation of the adjacent responsive gene(s) [80]. Because the
resulting increase in gene expression is directly proportional to the toxicity of the
Table 3 Eukaryotic cell-based bioluminescent bioreporters for endocrine-disrupting chemicals and recent environmental applications
Host cell Reporter construct (reporter name) Detection limit EC50 References Recent environmental application
(organism)
Estrogenic and antiestrogenic
S. cerevisiae hERa, ERE(92)-luxCDABE (BLYES) 45 pM E2 0.24 nM E2 [118] Freshwater [143]
Drinking water [143, 144]
S. cerevisiae hERa, ERE(92)-luc (BMAEREluc/ 30 pM E2 0.5 nM E2 [116] Wastewater [145]
ERa)
S. cerevisiae hERb, ERE-luc 0.1 nM E2 0.5 nM E2 [116] N/Aa
(BMAEREluc/ERb)
MCF-7 ERE-luc (MVLN) 1 pM 20 pM [98, 99] River water [137]
(human) Sediment [137]
Wastewater [146]
MCF-7 ERE-luc (MELN) 1 pM E2 5 pM E2 [102] Wastewater [125, 147]
(human) Freshwater [148, 149]
Sediment [148, 149]
T-47D (human) ERE(93)-luc (T-47D ER-CALUX) 0.5 pM E2 6 pM E2 [104] Wastewater [150, 151]
Freshwater [151, 152]
Sediment [151]
T-47D (human) ERE(93)-luc (T-47D-KBluc) 1 pM E2 10 pM E2 [105] Wastewater [126, 153, 154]
Freshwater [153]
HeLa (human) hERa, ERE-luc (HELNa) 1 pM E2 5 pM E2 [102] Drinking water [155]
HeLa (human) hERb, ERE-luc (HELNb) 1 pM E2 10 pM E2 [102] N/A
U2-OS (human) hERa, ERE(93)-luc (ERa-CALUX) 0.8 pM E2 20 pM E2 [113] Indoor dust [156]
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays
Host cell Reporter construct (reporter name) Detection limit EC50 References Recent environmental application
(organism)
S. cerevisiae hAR, ARE(92)-luc (BMAAREluc/AR) 50 pM testosterone 10 nM [116, 117] Sediment [137]
testosterone
0.5 nM DHT 5.5 M DHT
T-47D (human) ARE(92)-luc (AR-LUX) 46 pM methyltrieno-lone 86 pM [107] Freshwater [158]
methyltrieno-
lone
MDA-MB-453 MMTV-luc (final reporter known as 0.1 nM DHT [108, 126] Wastewater [126, 159]
(human) MDA-kb20
O2-US (human) hAR, HRE (hormone response 3.6 pM DHT 0.13 nM DHT [113] Drinking water, wastewater, and
element)-luc freshwater [157]
Glucocorticoid receptor (GR) agonist and antagonist
MDA-MB-453 MMTV-luc (final reporter known as 10 nM dexamethasone N/A [108] Wastewater [159]
(human) MDA-kb2)
O2-US hGR, HRE(93)-luc (final reporter 0.2 pM dexamethasone 0.37 nM [113] Wastewater [160]
known as GR-CALUX) dexamethasone
Thyroid receptor (TR) agonist and antagonist
PC12 r(rat) Avian TRa1, TRE(9 4)-luc (PC-DR- 30 pM 3,3’,5-triiodo-L- 0.18 nM T3 [161] Wastewater, drinking water, and
LUC) thyronine (T3) surface water [162]
Progesterone receptor (PR) agonist and antagonist
O2-US hPR, HRE(93)-luc (PR-CALUX) 1.3 pM Org2058 0.09 nM Org2058 [113] Indoor dust [156]
Drinking water, wastewater, and
freshwater [157]
a
N/A not available
T. Xu et al.
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 133
binding chemical [81], this process provides an excellent platform for the devel-
opment of regulatable bioluminescent bioreporter-based dioxin detection
strategies.
The first dioxin-responsive bioluminescent reporter to be developed was used
by Postlind et al. [82] to track expression of the CYP1A1 and CYP1A2 genes of the
human cytochrome P450 gene family. This was accomplished by cloning the 50
flanking region from either the CYP1A1 or CYP1A2 genes upstream of a luc gene
in a human expression vector. These vectors were then introduced to a human
hepatoma (HepG2) cell line and challenged with TCDD and other compounds.
Under transient transfection conditions where cells were exposed to TCDD for
24 h the day after plasmid introduction, reporters expressing the CYP1A2 50
flanking sequences displayed a detection minimum of 0.01 nM TCDD, whereas
those expressing CYP1A1 50 sequences were detectable down to a concentration of
0.001 nM. Each was capable of responding to TCDD treatment in a dose–response
fashion, however, although the signal maximum for the CYP1A2-based reporter
was 10-fold over control upon treatment with 10-nM TCDD, the CYP1A1-based
reporter reached its signal maximum at 65-fold over control upon 100-nM treat-
ment. This gave the CYP1A1-based reporter both a larger range of detection, as
well as a greater signal intensity throughout that range. Commensurate with its
lackluster performance compared to CYP1A1, the CYP1A2-based reporter con-
struct was not able to function at all under stable expression conditions. Whereas
the CYP1A1-based reporter was capable of stable expression and could detect
10-nM TCDD at 0.5 h postexposure, with increasing reporter activity positively
correlating with exposure time up to 24 h. The EC50 for TCDD detection by this
reporter was determined to be 0.35 nM, making it the first functionally useful
dioxin-detecting human cell line.
Building upon this expression strategy, Garrison et al. [80] used a construct
consisting of a mouse mammary tumor virus promoter mediated luc gene under the
control of a 484 base pair 50 upstream mouse Cyp1a1 gene sequence that contained
4 DREs to generate a range of bioluminescent dioxin-responsive bioreporter cell
lines. When treated with 1 nM TCDD for 24 h the day after plasmid introduction,
all of these lines were capable of responding to TCDD challenge with a
corresponding bioluminescent output. This demonstrated that the mouse DREs
could be successfully recognized by the dioxin:AhR:ARNT complex across a wide
range of species, and therefore that the assay could be performed in specific cell
types to determine species-specific bioavailability of dioxins and dioxin-like
compounds. Bolstered by these findings, a second set of stably transfected dioxin-
responsive bioluminescent bioreporter cell lines were developed. Of these cell
lines, the mouse H1L1.1c2 line (Hepa1-derived) was chosen as a model for
characterization because it had the greatest level of induction and was found to
respond reliably to TCDD treatment in a dose–response fashion (although this
dose–response relationship was also reported for the human HepG2-derived
HG2L1.1c3 cell line, no data were presented to support this claim at the time). The
model H1L1.1c2 reporter cell line displayed a minimal detection limit
between 0.1 and 1.0 pM of TCDD, with a maximal induction of 80-fold over
134 T. Xu et al.
control at 1.0 nM, providing an ED50 of 0.02 nM. This is less than the previous
value of 0.35 nM established by Postlind et al. [82] with their human HepG2 cell
line, and represented a lower maximal detection limit (1.0 vs. 100 nM) as well.
Also unlike the Postlind study, Garrison et al. [80] were not able to detect a
bioluminescent signal until 2 h posttreatment, and reached a maximum induction
at 4 h posttreatment following application of 1.0-nM TCDD. However, despite
being mostly in line with the Postlind study, the Garrison study remains notable for
its demonstration of bio-reporter function across a wide variety of cell lines, and
for demonstrating that the inducing chemicals did not act as competitive substrates
for the luciferase enzyme, providing a significant advantage for the bioluminescent
assay over older, more traditional chemical-based assays.
Following up these successful demonstrations against specific chemicals in
laboratory settings, Murk et al. [83] transported the system to a rat hepatoma
H4IIE cell line and rebranded its use as a chemical-activated luciferase expression
(CALUX) assay. Using this new cell line they were able to demonstrate TCDD
detection down to 0.5 fM, with a maximum detection limit between 100 pM and
1.0 nM, and an EC50 of 10 pM. Similar to the earlier experiments [80, 82], they
were also able to demonstrate a dose-response relationship between TCDD and
bioluminescent signal, and did not discover any significant substrate inhibition.
What solidified the CALUX assay as the predominant method for dioxin-like
compound detection, however, was the successful demonstration of its use with
environmental samples and its ability to mimic the results of existing, more com-
plex and more costly in vivo assay results [83]. Murk and colleagues demonstrated
that the luc-expressing H4IIE reporter cells could be exposed to extracted sediment
and water samples to determine toxic equivalency factors rapidly and inexpen-
sively. Although it was ultimately determined that unpurified sediment samples
could become toxic to the H4IIE-luc cells, it is primarily highlighted that purifi-
cation was not required for water samples, which significantly improved the speed
at which they could be assayed. Perhaps more important, however, was the
validation of the CALUX assay against the in vitro zebrafish early life stage assay.
Although the presence of heavy metals led to a poor correlation between the two
assays when performed directly in unprocessed environmental water samples, this
correlation significantly improved following sample extraction. When compared
side by side under laboratory controlled conditions, the EC50 as determined by the
zebrafish early life stage assay was found to be 21 pM, whereas the EC50 of the
CALUX assay was 10 pM. Moreover, the working range for the CALUX assay was
improved compared to the zebrafish early life stage assay, and reduced both the
time and cost involved in its performance.
Through its low cost, lack of substrate inhibition, ability to be adapted for
high-throughput, and ability to function in disparate cellular backgrounds, the
CALUX assay has become the predominant method for assaying dioxin and dioxin-
like chemicals using mammalian cells. Since its early demonstrations as a tool for
laboratory-based chemical toxicity testing and environmental pollutant screening, it
has been used for a wide variety of applications including veterinary [84], food
product testing [85–88], and human clinical sample analysis [89, 90]. And as the
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 135
applications for the CALUX assay have expanded, so have the organisms that have
been employed for its use. Although still predominantly performed in mammalian
cell lines, recently the assay has been reconstituted in yeast by co-expressing the
genes for AhR and ARNT with a DRE-mediated luc luciferase. This switch away
from human cells and into the microbial eukaryote S. cerevisiae was done with the
hope that it would provide a more robust and simplified expression system that
could improve deployability and reduce costs. It was found that the yeast-based
system could respond to TCDD treatment in a dose–response fashion, with a
minimum detection time of 3.5 h and an EC50 value similar to that of the early
human cellular reporters at 4.8 nM. Inasmuch as this assay was validated against the
classical H4IIE-luc cell line-based CALUX assay and found to be in good agree-
ment [91], it has since been used as a first stage in vivo screen for dioxin-like
chemical load detection in composted sewage sludge [91], proving its worth as an
alternative means of detection when human cell lines cannot be used.
As a final note, with the use of CALUX assays continuing to proliferate, it is
important to recognize that the results of the assay can vary from lab to lab, and
even from run to run [92]. In a validation study, Besselink et al. [93] found 14.6
and 26.1 % intralaboratory assay reliability levels for pure compounds and whole
matrix, respectively, and 6.5 and 27.9 % interlaboratory assay reliability levels for
pure compounds and whole matrix, respectively. Therefore, in light of these dis-
crepancies, it is important to review the sample cleanup methods, the effects of the
solvents used during extraction and testing, any known interaction with synergistic
or antagonistic compounds used, the cell line utilized, and the analysis methods
employed before comparing results between multiple sources in order to determine
the validity of the comparison [92]. Despite these caveats, however, the CALUX
assay remains the predominant method for bioluminescent screening of
compounds eliciting dioxin-like activities.
Moving away from the MCF-7 cell line, Legler et al. [104] developed a T-47D
cell-based ER-CALUX reporter cell line for detection of ER agonists and antag-
onists. Using a minimal TATA box promoter and three tandem repeats of ERE,
this cell line (T-47D ER-CALUX) displayed very low background biolumines-
cence in solvent controls and a maximum induction of approximately 100- and
76-fold compared to unexposed background following a 24-h exposure to 30 pM
E2 in 24- and 96-well plate formats, respectively. This bioassay was also capable
of detecting E2 down to *0.5 pM and is the most sensitive among reported
estrogen-specific assays. A similar T-47D-KBluc reporter developed by Wilson
et al. [105] has a comparable EC50 value for E2 (10 pM in KBluc vs. 6 pM in
EREtata-luc) but possesses a larger dynamic detection range from 1 pM to 100
nM. However, despite these similarities, it is worth noting that controversial
results were obtained in a follow-up study comparing the in vitro T-47D
ER-CALUX assay with an in vivo transgenic zebrafish assay expressing the same
reporter construct [106]. Despite the synthetic estrogen 17a-ethynylestradiol (EE2)
testing 100 times more potent than E2 in the transgenic zebrafish assay, it showed
equal estrogen agonistic activity compared to E2 in the cell-based in vitro assay.
One possible explanation for these differences is that the binding affinities of test
compounds may be different when interacting with ERs that have originated from
different species, which would explain the poor performance of human cell-based
assays to predict toxicokinetics in zebrafish models.
Under similar development strategies, bioluminescent bioreporters screening for
AR agonists and antagonists have been generated as well. These reporters take
advantage of AR-positive cell lines, such as T-47D and the human breast cancer cell
line MDA-MB-453 in order to supply the receptors needed for successful activation
of their chosen bioluminescent expression systems. Blankvoort et al. [107] were the
first to develop a stable T-47D AR-LUX cell line for androgenic and antiandrogenic
effects by using a rat probasin promoter-derived ARE-mediated luc reporter con-
struct. This reporter cell line was shown to be capable of detecting methyltrienolone
down to 46 pM after 24 h of exposure and detecting the environmentally relevant
antiandrogenic compounds 4,40 -DDE and (RS)-3-(3,5-dichlorophenyl)-5-methyl-5-
vinyloxazolidine-2,4-dione (vinclozoline) as well. However, because of the coex-
pression of other hormone receptors (such as ERa, ERb, and PR) in the T-47D
cellular background, there remains a possibility that nonspecific responses may have
been detected. To overcome this issue, Wilson et al. [108] developed a bioassay for
chemicals mimicking/blocking androgen and glucocorticoid activities utilizing the
MDA-MB-453 cell line, which expressed high levels of AR and GR but showed
undetectable or very low levels of alternative receptors [109, 110]. The resulting
reporter cell line, named MDA-kb2, expressed the luc gene under the regulation of
an AR- and GR-responsive mouse mammary tumor virus (MMTV) promoter and,
because of the characteristic low-level expression of competing hormone receptors,
provided a significant decrease in background activity that led to an increased
signal-to-noise detection ratio.
This decrease in alternative receptor expression resulting from the use of the
MDA-kb2 cell line helped to reduce cross-talk between different pathways [111],
138 T. Xu et al.
but was not the only means for accomplishing this goal. With the hope of providing
a more receptor-specific bioassay, several cell lines with little to undetectable levels
of nontarget receptors have been utilized through the introduction of specific
receptors that are not natively expressed. Examples of this approach include the use
of human cervical cancer cell line HeLa and osteosarcoma cell line U2-OS as
parental cells for reporter development. In addition to the MELN reporter cell line,
the HELNa and HELNb bioreporters were developed by Balaguer et al. [102] by
cointroducing ERa and ERb, respectively. Although the E2-generated biolumi-
nescent response of these cell lines is similar to that observed in MELN assays, it
should be noted that TCDD can elicit an antiestrogenic response in HeLa cell-based
assays compared to its demonstrated estrogenic activity in MELN assays. This
differential behavior highlights an example of the effect of pathway cross-talk (in
this case between the AhR- and ER-mediated pathways), which must always be
accounted for during data interpretation. To reduce the prevalence of this cross-talk,
a panel of CALUX bioassays has subsequently been developed for selective
detection of chemicals interacting with ERa, ERb, AR, GR, and PR using the U2-
OS cell line, which demonstrates little or no natural activity of any of these
receptors [112–114]. These reporter cell lines were generated by cointroducing a
vector that conferred constitutive receptor expression, and a second vector that
permitted target receptor-mediated expression of the luc gene. Using this approach,
the ERa- and AR-CALUX bioassays were also shown to be well correlated with
other animal-based assays (R2 value of 0.46 and 0.87 for AR and ERa assays,
respectively), making them useful tools for predicting potential in vivo activities
with a reduced chance of cross-talk–based interference [115].
For similar reasons to those listed above, as the techniques for genetic
expression continue to improve, there has been an increased interest in using the
lower eukaryotic organism S. cerevisiae as a platform for hormonally active
chemical screening. Due to their lack of human hormone receptor expression, fast
and robust growth, and relatively simplified genetic manipulation techniques,
yeast-based bioreporters can now be constructed using a stepwise transformation
of a recombinant human hormone receptor (e.g., hERa, hERb, and hAR) of
interest and a receptor-responsive reporter gene. Several luc-based yeast biore-
porters have been developed for the rapid profiling of estrogenic and androgenic
potentials, and have demonstrated a lower detection limit for E2 and dihydrotes-
tosterone of 30 and 50 pM, respectively [116, 117]. Despite this reduced sensi-
tivity compared to mammalian cell-line–based assays, the yeast-based assays were
capable of reporting relative potency of test chemicals more rapidly, with only a
2.5-h incubation time.
In particular, two yeast bioassays, BLYES [118] and BLYAS [119], which have
been developed to detect estrogenic and androgenic activity, respectively, stand
out from the other bioreporters mentioned above with respect to their choice of
bioluminescent reporter genes. Instead of using the luc reporter gene, each of these
utilizes a bioluminescent end point resulting from expression of the luxCDABE
genes. The switch to the lux system eliminates the need for exogenous luciferin
addition and/or cell lysis and permits autonomous bioluminescent signal
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 139
generation and detection. This allows the assay to proceed more rapidly, and with
near realtime signaling. Bioluminescent signal detection can occur as early as 1 h
after exposure to 2.8 nM E2 in the BLYES assay [118], which allowed the BLYES
and BLYAS assays to be used to evaluate the toxicity and potential endo-
crine-disrupting activities of a battery of 68 chemicals quickly and efficiently in a
cost-effective manner [120].
Compared to bioreporters relying on endogenous receptor-mediated signaling,
the test responses generated using recombinant yeast bioreporters and mammalian
bioreporters with manually introduced receptors are less likely to be subject to
nonspecific interactions, which may provide improved mechanistic insight.
However, it still remains to be seen if an enhanced prediction of in vivo effects
might be achieved using bioassays without exogenous manipulation of the
signaling receptors. This creates a potential tradeoff that will need to be evaluated
on a case-by-case basis.
4 Conclusions
surveillance and modeling of human/animal health impacts. This includes both the
lower eukaryotic yeast and mammalian cell lines that almost exclusively rely upon
firefly luciferase as a signaling element, although newer versions of ‘‘humanized’’
bacterial luciferase capable of being expressed under eukaryotic genetic controls
without the necessary addition of a light-activating substrate are becoming
available for higher throughput, more data intensive, realtime chemical toxicity
profiling [130]. As the inventory of bioluminescent bioreporters expands toward
more chemical targets with greater specificity, sensitivity, and human relevance, it
is clear that the bioreporter’s role as an environmental sentinel is here to stay.
References
1. Yeh BJ, Lim WA (2007) Synthetic biology: lessons from the history of synthetic organic
chemistry. Nat Chem Biol 3:521–525
2. Snyder R (2000) Overview of the toxicology of benzene. J Toxicol Env Health-Pt A
61:339–346
3. Dawson JJC, Iroegbu CO, Maciel H, Paton GI (2008) Application of luminescent biosensors
for monitoring the degradation and toxicity of BTEX compounds in soils. J Appl Microbiol
104:141–151
4. Girotti S, Bolelli L, Roda A, Gentilomi G, Musiani M (2002) Improved detection of toxic
chemicals using bioluminescent bacteria. Anal Chim Acta 471:113–120
5. Xu T, Close DM, Sayler GS, Ripp SA (2013) Genetically modified whole-cell bioreporters
for environmental assessment. Ecol Indic 28:125–141
6. Williams PA, Murray K (1974) Metabolism of benzoate and methylbenzoates by
Pseudomonas putida mt-2: evidence for existance of a TOL plasmid. J Bacteriol
120:416–423
7. Franklin FCH, Bagdasarian M, Bagdasarian MM, Timmis KN (1981) Molecular and
functional analysis of the TOL Plasmid pWWO from Pseudomonas putida and cloning of
genes for the enitre regulated aromatic ring meta-cleavage pathway. Proc Natl Acad Sci
USA 78:7458–7462
8. Worsey MJ, Franklin FCH, Williams PA (1978) Regulation of degradative pathway
enzymes coded for by TOL plasmid pWWO from Pseudomonas putida mt-2. J Bacteriol
134:757–764
9. Worsey MJ, Williams PA (1975) Metabolism of toluene and xylenes by Pseudomonas
putida mt-2: evidence for a new function of TOL plasmid. J Bacteriol 124:7–13
10. Ramos JL, Marques S, Timmis KN (1997) Transcriptional control of the Pseudomonas tol
plasmid catabolic operons is achieved through an interplay of host factors and plasmid-
encoded regulators. An Rev Microbiol 51:341–373
11. Li Y-F, Li F-Y, Ho C-L, Liao VH-C (2008) Construction and comparison of fluorescence
and bioluminescence bacterial biosensors for the detection of bioavailable toluene and
related compounds. Environ Pollut 152:123–129
12. Urbanczyk H, Ast JC, Higgins MJ, Carson J, Dunlap PV (2007) Reclassification of Vibrio
fischeri, Vibrio logei, Vibrio salmonicida and Vibrio wodanis as Aliivibrio fischeri gen.
nov., comb. nov., Aliivibrio logei comb. nov., Aliivibrio salmonicida comb. nov and
Aliivibrio wodanis comb. nov. Int J Syst Evol Microbiol 57:2823–2829
13. Willardson BM, Wilkins JF, Rand TA, Schupp JM, Hill KK, Keim P, Jackson PJ (1998)
Development and testing of a bacterial biosensor for toluene-based environmental
contaminants. Appl Environ Microbiol 64:1006–1012
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 143
14. Tecon R, Beggah S, Czechowska K, Sentchilo V, Chronopoulou PM, McGenity TJ, van der
Meer JR (2010) Development of a multistrain bacterial bioreporter platform for the
monitoring of hydrocarbon contaminants in marine environments. Environ Sci Technol
44:1049–1055
15. Applegate BM, Kehrmeyer SR, Sayler GS (1998) A chromosomally based tod-luxCDABE
whole-cell reporter for benzene, toluene, ethybenzene, and xylene (BTEX) sensing. Appl
Environ Microbiol 64:2730–2735
16. Zylstra G, McCombie W, Gibson D, Finette B (1988) Toluene degradation by Pseudomonas
putida F1: genetic organization of the tod operon. Appl Environ Microbiol 54:1498–1503
17. Wang Y, Rawlings M, Gibson DT, Labbe D, Bergeron H, Brousseau R, Lau PCK (1995)
Identification of a membrane protein and a truncated LysR type regulator associated with
the toluene degradation pathway in Pseudomonas putida F1. Mol Gen Genet 246:570–579
18. Kuncova G, Pazlarova J, Hlavata A, Ripp S, Sayler GS (2011) Bioluminescent bioreporter
Pseudomonas putida TVA8 as a detector of water pollution. Operational conditions and
selectivity of free cells sensor. Ecol Indic 11:882–887
19. Stiner L, Halverson LJ (2002) Development and characterization of a green fluorescent
protein-based bacterial biosensor for bioavailable toluene and related compounds. Appl
Environ Microbiol 68:1962–1971
20. Bhattacharyya J, Read D, Amos S, Dooley S, Killham K, Paton GI (2005) Biosensor-based
diagnostics of contaminated groundwater: assessment and remediation strategy. Environ
Pollut 134:485–492
21. Eaton RW, Timmis KN (1986) Characterization of a plasmid-specified pathway for
catabolism of isopropylbenzne in Pseudomonas putida RE204. J Bacteriol 168:123–131
22. Selifonova OV, Eaton RW (1996) Use of an ipb-lux fusion to study regulation of the
isopropylbenzene catabolism operon of Pseudomonas putida RE204 and to detect
hydrophobic pollutants in the environment. Appl Environ Microbiol 62:778–783
23. Mumtaz MM, George JD, Gold KW, Cibulas W, Derosa CT (1996) ATSDR evaluation of
health effects of chemicals. 4. Polycyclic aromatic hydrocarbons (PAHs): understanding a
complex problem. Toxicol Ind Health 12:742–971
24. Grund AD, Gunsalus IC (1983) Cloning of genes for naphthalene metabolism in
Pseudomonas putida. J Bacteriol 156:89–94
25. Burlage RS, Sayler GS, Larimer F (1990) Monitoring of naphthalene catabolism by
bioluminescence with nah-lux transcriptional fusions. J Bacteriol 172:4749–4757
26. Dorn JG, Brusseau ML, Maier RM (2005) Real-time, in situ monitoring of bioactive zone
dynamics in heterogeneous systems. Environ Sci Technol 39:8898–8905
27. Dorn JG, Frye RJ, Maier RM (2003) Effect of temperature, pH, and initial cell number on
luxCDABE and nah gene expression during naphthalene and salicylate catabolism in the
bioreporter organism Pseudomonas putida RB1353. Appl Environ Microbiol 69:2209–2216
28. Dorn JG, Mahal MK, Brusseau ML, Maier RM (2004) Employing a novel fiber optic
detection system to monitor the dynamics of in situ lux bioreporter activity in porous media:
system performance update. Anal Chim Acta 525:63–74
29. King JMH, Digrazia PM, Applegate B, Burlage R, Sanseverino J, Dunbar P, Larimer F,
Sayler GS (1990) Rapid, sensitive bioluminescent reporter technology for naphthalene
exposure and biodegradation. Science 249:778–781
30. Trogl J, Chauhan A, Ripp S, Layton AC, Kuncova G, Sayler GS (2012) Pseudomonas
fluorescens HK44: lessons learned from a model whole-cell bioreporter with a broad
application history. Sensors 12:1544–1571
31. Valdman E, Gutz IGR (2008) Bioluminescent sensor for naphthalene in air: Cell
immobilization and evaluation with a dynamic standard atmosphere generator. Sens
Actuator B-Chem 133:656–663
32. Ripp S, Nivens DE, Ahn Y, Werner C, Jarrell J, Easter JP, Cox CD, Burlage RS, Sayler GS
(2000) Controlled field release of a bioluminescent genetically engineered microorganism
for bioremediation process monitoring and control. Environ Sci Technol 34:846–853
144 T. Xu et al.
33. Sayler GS, Ripp S (2000) Field applications of genetically engineered microorganisms for
bioremediation processes. Curr Opin Biotechnol 11:286–289
34. Laurie AD, Lloyd-Jones G (1999) The phn genes of Burkholderia sp. strain RP007
constitute a divergent gene cluster for polycyclic aromatic hydrocarbon catabolism.
J Bacteriol 181:531–540
35. Chakrabarty A, Chou G, Gunsalus I (1973) Genetic regulation of octane dissimilation
plasmid in Pseudomonas. Proc Natl Acad Sci USA 70:1137–1140
36. Eggink G, Engel H, Meijer W, Otten J, Kingma J, Witholt B (1988) Alkane utilization in
Pseudomonas oleovorans. Structure and function of the regulatory locus alkR. J Biol Chem
263:13400–13405
37. Sticher P, Jaspers MCM, Stemmler K, Harms H, Zehnder AJB, van der Meer JR (1997)
Development and characterization of a whole-cell bioluminescent sensor for bioavailable
middle-chain alkanes in contaminated groundwater samples. Appl Environ Microbiol
63:4053–4060
38. Owen DJ, Eggink G, Hauer B, Kok M, McBeth DL, Yang YL, Shapiro JA (1984) Physical
structure, genetic content and expression of the alkBAC operon. Mol Gen Genet
197:373–383
39. Minak-Bernero V, Bare RE, Haith CE, Grossman MJ (2004) Detection of alkanes, alcohols,
and aldehydes using bioluminescence. Biotechnol Bioeng 87:170–177
40. Bosetti A, van Beilen JB, Preusting H, Lageveen RG, Witholt B (1992) Production of
primary aliphatic alcohols with a recombinant Pseudomonas strain, encoding the alkane
hydroxylase enzyme system. Enzyme Microb Technol 14:702–708
41. Francisco W, Abu-Soud H, Baldwin T, Raushel F (1993) Interaction of aldehyde substrate
and inhibitors to bacterial luciferase. J Biol Chem 268:24734–24741
42. Atlas RM, Hazen TC (2011) Oil biodegradation and bioremediation: a tale of the two worst
spills in U.S. history. Environ Sci Technol 45:6709–6715
43. Zhang D, He Y, Wang Y, Wang H, Wu L, Aries E, Huang WE (2012) Whole-cell bacterial
bioreporter for actively searching and sensing of alkanes and oil spills. Microb Biotechnol
5:87–97
44. Zhang DY, Fakhrullin RF, Ozmen M, Wang H, Wang J, Paunov VN, Li GH, Huang WE
(2011) Functionalization of whole-cell bacterial reporters with magnetic nanoparticles.
Microb Biotechnol 4:89–97
45. Kumari R, Tecon R, Beggah S, Rutler R, Arey JS, van der Meer JR (2011) Development of
bioreporter assays for the detection of bioavailability of long-chain alkanes based on the
marine bacterium Alcanivorax borkumensis strain SK2. Environ Microbiol 13:2808–2819
46. van Hylckama Vlieg JET, Janssen DB (2001) Formation and detoxification of reactive
intermediates in the metabolism of chlorinated ethenes. J Biotechnol 85:81–102
47. Arcangeli J-P, Arvin E (1997) Modeling of the cometabolic biodegradation of
trichloroethylene by toluene-oxidizing bacteria in a biofilm system. Environ Sci Technol
31:3044–3052
48. Sponza DT (2003) Toxicity and treatability of carbontetrachloride and tetrachloroethylene
in anaerobic batch cultures. Int Biodeterior Biodegrad 51:119–127
49. Shingleton JT, Applegate BM, Nagel AC, Bienkowski PR, Sayler GS (1998) Induction of
the tod operon by trichloroethylene in Pseudomonas putida TVA8. Appl Environ Microbiol
64:5049–5052
50. Phoenix P, Keane A, Patel A, Bergeron H, Ghoshal S, Lau P (2003) Characterization of a
new solvent-responsive gene locus in Pseudomonas putida F1 and its functionalization as a
versatile biosensor. Environ Microbiol 5:1309–1327
51. Lopes N, Hawkins SA, Jegier P, Menn F-M, Sayler GS, Ripp S (2012) Detection of
dichloromethane with a bioluminescent (lux) bacterial bioreporter. J Ind Microbiol
Biotechnol 39:45–53
52. Furukawa K, Fujihara H (2008) Microbial degradation of polychlorinated biphenyls:
Biochemical and molecular features. J Biosci Bioeng 105:433–449
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 145
53. Layton AC, Muccini M, Ghosh MM, Sayler GS (1998) Construction of a bioluminescent
reporter strain to detect polychlorinated biphenyls. Appl Environ Microbiol 64:5023–5026
54. Bradley C, Berube PR (2008) Characterization of anionic surfactant-induced toxicity in a
primary effluent. J Environ Eng Sci 7:63–70
55. Layton AC, Gregory B, Schultz TW, Sayler GS (1999) Validation of genetically engineered
bioluminescent surfactant resistant bacteria as toxicity assessment tools. Ecotox Environ
Safe 43:222–228
56. Park SH, Lee K, Chae JC, Kim CK (2004) Construction of transformant reporters carrying
fused genes using pcbC promoter of Pseudomonas sp DJ-12 for detection of aromatic
pollutants. Environ Monit Assess 92:241–251
57. Jaspers MCM, Suske WA, Schmid A, Goslings DAM, Kohler HPE, van der Meer JR (2000)
HbpR, a new member of the XylR/DmpR subclass within the NtrC family of bacterial
transcriptional activators, regulates expression of 2-hydroxybiphenyl metabolism in
Pseudomonas azelaica HBP1. J Bacteriol 182:405–417
58. Turner K, Xu S, Pasini P, Deo S, Bachas L, Daunert S (2007) Hydroxylated polychlorinated
biphenyl detection based on a genetically engineered bioluminescent whole-cell sensing
system. Anal Chem 79:5740–5745
59. Tropel D, Bahler A, Globig K, van der Meer JR (2004) Design of new promoters and of a
dual-bioreporter based on cross-activation by the two regulatory proteins XylR and HbpR.
Environ Microbiol 6:1186–1196
60. Krastanov A, Alexieva Z, Yemendzhiev H (2013) Microbial degradation of phenol and
phenolic derivatives. Eng Life Sci 13:76–87
61. Shingler V, Franklin FCH, Tsuda M, Holroyd D, Bagdasarian M (1989) Molecular analysis
of a plasmid-encoded phenol hydroxylase from Pseudomonas CF600. J Gen Microbiol
135:1083–1092
62. Shingler V, Bartilson M, Moore T (1993) Cloning and nucleotide-sequencing of the gene
encoding the positive regulator (DmpR) of the phenol catabolic pathway encoded by
PVI150 and identification of DmpR as a member of the NtrC family of transcriptional
activators. J Bacteriol 175:1596–1604
63. Leedjarv A, Ivask A, Virta M, Kahru A (2006) Analysis of bioavailable phenols from
natural samples by recombinant luminescent bacterial sensors. Chemosphere 64:1910–1919
64. Wise AA, Kuske CR (2000) Generation of novel bacterial regulatory proteins that detect
priority pollutant phenols. Appl Environ Microbiol 66:163–169
65. Gupta S, Saxena M, Saini N, Mahmooduzzafar, Kumar R, Kumar A (2012) An effective
strategy for a whole-cell biosensor based on putative effector interaction site of the
regulatory DmpR protein. PLoS ONE 7: e43527
66. Ehrt S, Schirmer F, Hillen W (1995) Genetic organization, nucleotide sequence and
regulation of expression of genes encoding phenol hydroxylase and catechol 1,2-
dioxygenase in Acinetobacter calcoaceticus NCIB8250. Mol Microbiol 18:13–20
67. Abd-El-Haleem D, Ripp S, Scott C, Sayler GS (2002) A luxCDABE-based bioluminescent
bioreporter for the detection of phenol. J Ind Microbiol Biotechnol 29:233–237
68. Zaki S, Abd-El-Haleem D, Abulhamd A, Elbery H, AbuElreesh G (2008) Influence of
phenolics on the sensitivity of free and immobilized bioluminescent Acinetobacter
bacterium. Microbiol Res 163:277–285
69. Wiles S, Whiteley AS, Philp JC, Bailey MJ (2003) Development of bespoke bioluminescent
reporters with the potential for in situ deployment within a phenolic-remediating wastewater
treatment system. J Microbiol Methods 55:667–677
70. Ghosh SK, Doctor PB (1992) Toxicity screening of phenol using Microtox. Environ Toxicol
Water Quality 7:157–163
71. Ismailov AD, Pogosyan SI, Mitrofanova TI, Egorov NS, Netrusov AI (2000) Bacterial
bioluminescence inhibition by chlorophenols. Appl Biochem Microbiol 36:404–408
72. Kudryasheva N, Vetrova E, Kuznetsov A, Kratasyuk V, Stom D (2002) Bioluminescence
assays: Effects of quinones and phenols. Ecotox Environ Safe 53:221–225
146 T. Xu et al.
73. Berglind R, Leffler P, Sjostrom M (2010) Interactions between pH, potassium, calcium,
bromide, and phenol and their effects on the bioluminescence of Vibrio fischeri. J Toxicol
Env Health-Pt A 73:1102–1112
74. Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser R, Prins GS, Soto AM,
Zoeller RT, Gore AC (2009) Endocrine-disrupting chemicals: an endocrine society
scientific statement. Endocr Rev 30:293–342
75. Alcock RE, Behnisch PA, Jones KC, Hagenmaier H (1998) Dioxin-like PCBs in the
environment—human exposure and the significance of sources. Chemosphere
37:1457–1472
76. Soto AM, Sonnenschein C, Chung KL, Fernandez MF, Olea N, Serrano FO (1995) The E-
SCREEN assay as a tool to identify estrogens—an update on estrogenic environmental
pollutants. Environ Health Perspect 103:113–122
77. Van den Berg M, Birnbaum L, Bosveld A, Brunström B, Cook P, Feeley M, Giesy JP,
Hanberg A, Hasegawa R, Kennedy SW (1998) Toxic equivalency factors (TEFs) for PCBs,
PCDDs, PCDFs for humans and wildlife. Environ Health Perspect 106:775–792
78. Ahlborg UG, Brouwer A, Fingerhut MA, Jacobson JL, Jacobson SW, Kennedy SW, Kettrup
AA, Koeman JH, Poiger H, Rappe C (1992) Impact of polychlorinated dibenzo-p-dioxins,
dibenzofurans, and biphenyls on human and environmental health, with special emphasis on
application of the toxic equivalency factor concept. Environ Toxicol Pharmacol
228:179–199
79. Peterson RE, Theobald HM, Kimmel GL (1993) Developmental and reproductive toxicity
of dioxins and related compounds: cross-species comparisons. Crit Rev Toxicol 23:283–335
80. Garrison P, Tullis K, Aarts J, Brouwer A, Giesy J, Denison M (1996) Species-specific
recombinant cell lines as bioassay systems for the detection of 2,3,7,8-tetrachlorodibenzo-p-
dioxin-like chemicals. Toxicol Sci 30:194–203
81. Safe SH (1995) Modulation of gene expression and endocrine response pathways by 2,3,7,8-
tetrachlorodibenzo-p-dioxin and related compounds. Pharmacol Ther 67:247–281
82. Postlind H, Vu T, Tukey R, Quattrochi LC (1993) Response of human CYP1-luciferase
plasmids to 2,3,7,8-tetrachlorodibenzo-p-dioxin and polycyclic aromatic hydrocarbons.
Toxicol Appl Pharmacol 118:255–262
83. Murk AJ, Legler J, Denison MS, Giesy JP, vandeGuchte C, Brouwer A (1996) Chemical-
activated luciferase gene expression (CALUX): a novel in vitro bioassay for Ah receptor
active compounds in sediments and pore water. Fundam Appl Toxicol 33:149–160
84. Murk AJ, Leonards PEG, Bulder AS, Jonas AS, Rozemeijer MJC, Denison MS, Koeman
JH, Brouwer A (1997) The CALUX (chemical-activated luciferase expression) assay
adapted and validated for measuring TCDD equivalents in blood plasma. Environ Toxicol
Chem 16:1583–1589
85. Bovee TFH, Hoogenboom LAP, Hamers ARM, Traag WA, Zuidema T, Aarts J, Brouwer A,
Kuiper HA (1998) Validation and use of the CALUX-bioassay for the determination of
dioxins and PCBs in bovine milk. Food Addit Contam 15:863–875
86. Cederberg T, Laier P, Vinggaard AM (2002) Screening of food samples for dioxin levels:
comparison of GC-MS determination with the CALUX bioassay. Organohalogen Compd
58:409–412
87. Tsutsumi T, Amakura Y, Nakamura M, Brown DJ, Clark GC, Sasaki K, Toyoda M, Maitani
T (2003) Validation of the CALUX bioassay for the screening of PCDD/Fs and dioxin-like
PCBs in retail fish. Analyst 128:486–492
88. Van Overmeire I, Carbonnelle S, Van Loco J, Roos P, Brown D, Chu M, Clark G, Goeyens
L (2002) Validation of the CALUX bioassay: quantitative screening approach.
Organohalogen Compd 58:353–356
89. Pauwels A, Cenijn PH, Schepens P, Brouwer A (2000) Comparison of chemical-activated
luciferase gene expression bioassay and gas chromatography for PCB determination in
human serum and follicular fluid. Environ Health Perspect 108:553–557
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 147
90. Van Wouwe N, Windal I, Vanderperren H, Eppe G, Xhrouet C, Massart A-C, Debacker N,
Sasse A, Baeyens W, De Pauw E (2004) Validation of the CALUX bioassay for PCDD/F
analyses in human blood plasma and comparison with GC-HRMS. Talanta 63:1157–1167
91. Leskinen P, Hilscherova K, Sidlova T, Kiviranta H, Pessala P, Salo S, Verta M, Virta M
(2008) Detecting AhR ligands in sediments using bioluminescent reporter yeast. Biosens
Bioelectron 23:1850–1855
92. Windal I, Denison MS, Birnbaum LS, Van Wouwe N, Baeyens W, Goeyens L (2005)
Chemically activated luciferase gene expression (CALUX) cell bioassay analysis for the
estimation of dioxin-like activity: critical parameters of the CALUX procedure that impact
assay results. Environ Sci Technol 39:7357–7364
93. Besselink HT, Schipper C, Klamer H, Leonards P, Verhaar H, Felzel E, Murk AJ, Thain J,
Hosoe K, Schoeters G, Legler J, Brouwer B (2004) Intra- and interlaboratory calibration of
Ò
the DR CALUX bioassay for the analysis of dioxins and dioxin-like chemicals in
sediments. Environ Toxicol Chem 23:2781–2789
94. Colborn T, vom Saal FS, Soto AM (1993) Developmental effects of endocrine-disrupting
chemicals in wildlife and humans. Environ Health Perspect 101:378–384
95. Kavlock RJ, Daston GP, DeRosa C, FennerCrisp P, Gray LE, Kaattari S, Lucier G, Luster
M, Mac MJ, Maczka C, Miller R, Moore J, Rolland R, Scott G, Sheehan DM, Sinks T,
Tilson HA (1996) Research needs for the risk assessment of health and environmental
effects of endocrine disruptors: a report of the US EPA-sponsored workshop. Environ
Health Perspect 104:715–740
96. Eltzov E, Kushmaro A, Marks RS (2009) Biosensors for endocrine disruptors. In: Shaw I
(eds) Endocrine-disrupting chemicals in food. Woodhead Publishing in Food Science
Technology and Nutrition, pp 183–208
97. Svobodova K, Cajthaml T (2010) New in vitro reporter gene bioassays for screening of
hormonal active compounds in the environment. Appl Microbiol Biotechnol 88:839–847
98. Pons M, Gagne D, Nicolas JC, Mehtali M (1990) A new cellular model of response to
estrogens: a bioluminescent test to characterize (anti)estrogen molecules. Biotechniques
9:450
99. Demirpence E, Duchesne MJ, Badia E, Gagne D, Pons M (1993) MVLN cells—a
bioluminescent MCF-7-derived cell line to study the modulation of estrogenic activity.
J Steroid Biochem Mol Biol 46:355–364
100. Shue MF, Chen FA, Chen TC (2010) Total estrogenic activity and nonylphenol
concentration in the Donggang River. Taiwan. Environ Monit Assess 168:91–101
101. Wang C, Wang T, Liu W, Ruan T, Zhou QF, Liu JY, Zhang AQ, Zhao B, Jiang GB (2012)
The in vitro estrogenic activities of polyfluorinated iodine alkanes. Environ Health Perspect
120:119–125
102. Balaguer P, Francois F, Comunale F, Fenet H, Boussioux AM, Pons M, Nicolas JC, Casellas
C (1999) Reporter cell lines to study the estrogenic effects of xenoestrogens. Sci Total
Environ 233:47–56
103. Witters H, Freyberger A, Smits K, Vangenechten C, Lofink W, Weimer M, Bremer S, Ahr
PHJ, Berckmans P (2010) The assessment of estrogenic or anti-estrogenic activity of
chemicals by the human stably transfected estrogen sensitive MELN cell line: results of test
performance and transferability. Reprod Toxicol 30:60–72
104. Legler J, van den Brink CE, Brouwer A, Murk AJ, van der Saag PT, Vethaak AD, van der
Burg B (1999) Development of a stably transfected estrogen receptor-mediated luciferase
reporter gene assay in the human T47D breast cancer cell line. Toxicol Sci 48:55–66
105. Wilson VS, Bobseine K, Gray LE (2004) Development and characterization of a cell line
that stably expresses an estrogen-responsive luciferase reporter for the detection of estrogen
receptor agonist and antagonists. Toxicol Sci 81:69–77
106. Legler J, Zeinstra LM, Schuitemaker F, Lanser PH, Bogerd J, Brouwer A, Vethaak AD, De
Voogt P, Murk AJ, Van der Burg B (2002) Comparison of in vivo and in vitro reporter gene
assays for short-term screening of estrogenic activity. Environ Sci Technol 36:4410–4415
148 T. Xu et al.
107. Blankvoort BMG, de Groene EM, van Meeteren-Kreikamp AP, Witkamp RF, Rodenburg
RJT, Aarts J (2001) Development of an androgen reporter gene assay (AR-LUX) utilizing a
human cell line with an endogenously regulated androgen receptor. Anal Biochem
298:93–102
108. Wilson VS, Bobseine K, Lambright CR, Gray LE (2002) A novel cell line, MDA-kb2, that
stably expresses an androgen- and glucocorticoid-responsive reporter for the detection of
hormone receptor agonists and antagonists. Toxicol Sci 66:69–81
109. Hall RE, Tilley WD, McPhaul MJ, Sutherland RL (1992) Regulation of androgen receptor-
gene expression by steroids and retinoic acid in human breast-cancer cells. Int J Cancer
52:778–784
110. Vladusic EA, Hornby AE, Guerra-Vladusic FK, Lakins J, Lupu R (2000) Expression and
regulation of estrogen receptor beta in human breast tumors and cell lines. Oncol Rep
7:157–167
111. Aranda A, Pascual A (2001) Nuclear hormone receptors and gene expression. Physiol Rev
81:1269–1304
112. Quaedackers ME, Van den Brink CE, Wissink S, Schreurs R, Gustafsson JA, Van der Saag
PT, Van der Burg B (2001) 4-hydroxytamoxifen trans-represses nuclear factor-kappa B
activity in human osteoblastic U2-OS cells through estrogen receptor (ER)alpha, and not
through ER beta. Endocrinology 142:1156–1166
113. Sonneveld E, Jansen HJ, Riteco JAC, Brouwer A, van der Burg B (2005) Development of
androgen- and estrogen-responsive bioassays, members of a panel of human cell line-based
highly selective steroid-responsive bioassays. Toxicol Sci 83:136–148
114. van der Burg B, Schreurs R, van der Linden S, Seinen W, Brouwer A, Sonneveld E (2008)
Endocrine effects of polycyclic musks: do we smell a rat? Int J Androl 31:188–193
115. Sonneveld E, Riteco JAC, Jansen HJ, Pieterse B, Brouwer A, Schoonen WG, van der Burg
B (2006) Comparison of in vitro and in vivo screening models for androgenic and estrogenic
activities. Toxicol Sci 89:173–187
116. Leskinen P, Michelini E, Picard D, Karp M, Virta M (2005) Bioluminescent yeast assays for
detecting estrogenic and androgenic activity in different matrices. Chemosphere 61:259–266
117. Michelini E, Leskinen P, Virta M, Karp M, Roda A (2005) A new recombinant cell-based
bioluminescent assay for sensitive androgen-like compound detection. Biosens Bioelectron
20:2261–2267
118. Sanseverino J, Gupta RK, Layton AC, Patterson SS, Ripp SA, Saidak L, Simpson ML,
Schultz TW, Sayler GS (2005) Use of Saccharomyces cerevisiae BLYES expressing
bacterial bioluminescence for rapid, sensitive detection of estrogenic compounds. Appl
Environ Microbiol 71:4455–4460
119. Eldridge ML, Sanseverino J, Layton AC, Easter JP, Schultz TW, Sayler GS (2007)
Saccharomyces cerevisiae BLYAS, a new bioluminescent bioreporter for detection of
androgenic compounds. Appl Environ Microbiol 73:6012–6018
120. Sanseverino J, Eldridge ML, Layton AC, Easter JP, Yarbrough J, Schultz TW, Sayler GS
(2009) Screening of potentially hormonally active chemicals using bioluminescent yeast
bioreporters. Toxicol Sci 107:122–134
121. Vandenberg LN, Maffini MV, Sonnenschein C, Rubin BS, Soto AM (2009) Bisphenol-A
and the great divide: A review of controversies in the field of endocrine disruption. Endocr
Rev 30:75–95
122. Bonefeld-Jorgensen EC, Long MH, Hofmeister MV, Vinggaard AM (2007) Endocrine-
disrupting potential of bisphenol A, bisphenol A dimethacrylate, 4-n-nonylphenol, and
4-n-octylphenol in vitro: new data and a brief review. Environ Health Perspect 115:69–76
123. Mankidy R, Wiseman S, Ma H, Giesy JP (2013) Biological impact of phthalates. Toxicol
Lett 217:50–58
124. Preuss TG, Gurer-Orhan H, Meerman J, Ratte HT (2010) Some nonylphenol isomers show
antiestrogenic potency in the MVLN cell assay. Toxicol in Vitro 24:129–134
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 149
125. Schiliro T, Porfido A, Spina F, Varese GC, Gilli G (2012) Oestrogenic activity of a textile
industrial wastewater treatment plant effluent evaluated by the E-screen test and MELN
gene-reporter luciferase assay. Sci Total Environ 432:389–395
126. He YH, Wiseman SB, Hecker M, Zhang XW, Wang N, Perez LA, Jones PD, El-Din MG,
Martin JW, Giesy JP (2011) Effect of ozonation on the estrogenicity and androgenicity of
oil sands process-affected water. Environ Sci Technol 45:6268–6274
127. Pereira RO, Postigo C, de Alda ML, Daniel LA, Barcelo D (2011) Removal of estrogens
through water disinfection processes and formation of by-products. Chemosphere
82:789–799
128. Kortenkamp A (2007) Ten years of mixing cocktails: a review of combination effects of
endocrine-disrupting chemicals. Environ Health Perspect 115:98–105
129. Fenet H, Gomez E, Pillon A, Rosain D, Nicolas JC, Casellas C, Balaguer P (2003)
Estrogenic activity in water and sediments of a French river: contribution of alkylphenols.
Arch Environ Contam Toxicol 44:1–6
130. Close DM, Patterson SS, Ripp SA, Baek SJ, Sanseverino J, Sayler GS (2010) Autonomous
bioluminescent expression of the bacterial luciferase gene cassette (lux) in a mammalian
cell line. PLoS ONE 5:e12441
131. Bundy JG, Campbell CD, Paton GI (2001) Comparison of response of six different
luminescent bacterial bioassays to bioremediation of five contrasting oils. J Environ Monit
3:404–410
132. Diplock EE, Mardlin DP, Killham KS, Paton GI (2009) Predicting bioremediation of
hydrocarbons: Laboratory to field scale. Environ Pollut 157:1831–1840
133. Werlen C, Jaspers MCM, van der Meer JR (2004) Measurement of biologically available
naphthalene in gas and aqueous phases by use of a Pseudomonas putida biosensor. Appl
Environ Microbiol 70:43–51
134. Paton GI, Reid BJ, Sempled KT (2009) Application of a luminescence-based biosensor for
assessing naphthalene biodegradation in soils from a manufactured gas plant. Environ Pollut
157:1643–1648
135. Kapanen A, Vikman M, Rajasärkkä J, Virta M, Itävaara M (2013) Biotests for
environmental quality assessment of composted sewage sludge. Waste Manag
33:1451–1460
136. Sakai S, Takigami H (2003) Integrated biomonitoring of dioxin-like compounds for waste
management and environment. Ind Health 41:205–214
137. Hilscherova K, Dusek L, Sidlova T, Jalova V, Cupr P, Giesy JP, Nehyba S, Jarkovsky J,
Klanova J, Holoubek I (2010) Seasonally and regionally determined indication potential of
bioassays in contaminated river sediments. Environ Toxicol Chem 29:522–534
138. Kanematsu M, Hayashi A, Denison MS, Young TM (2009) Characterization and potential
environmental risks of leachate from shredded rubber mulches. Chemosphere 76:952–958
139. Richter CA, Tieber VL, Denison MS, Giesy JP (1997) An in vitro rainbow trout cell
bioassay for aryl hydrocarbon receptor-mediated toxins. Environ Toxicol Chem 16:543–550
140. Hahn ME (2002) Biomarkers and bioassays for detecting dioxin-like compounds in the
marine environment. Sci Total Environ 289:49–69
141. Yang J-H, Lee H-G, Park K-Y (2008) Development of human dermal epithelial cell-based
bioassay for the dioxins. Chemosphere 72:1188–1192
142. He G, Tsutsumi T, Zhao B, Baston DS, Zhao J, Heath-Pagliuso S, Denison MS (2011)
Third-generation Ah receptor–responsive luciferase reporter plasmids: Amplification of
dioxin-responsive elements dramatically increases CALUX bioassay sensitivity and
responsiveness. Toxicol Sci 123:511–522
143. Bergamasco AMD, Eldridge M, Sanseverino J, Sodre FF, Montagner CC, Pescara IC,
Jardim WF, Umbuzeiro GD (2011) Bioluminescent yeast estrogen assay (BLYES) as a
sensitive tool to monitor surface and drinking water for estrogenicity. J Environ Monit
13:3288–3293
150 T. Xu et al.
144. Jardim WF, Montagner CC, Pescara IC, Umbuzeiro GA, Bergamasco AMD, Eldridge ML,
Sodre FF (2012) An integrated approach to evaluate emerging contaminants in drinking
water. Sep Purif Technol 84:3–8
145. Salste L, Leskinen P, Virta M, Kronberg L (2007) Determination of estrogens and
estrogenic activity in wastewater effluent by chemical analysis and the bioluminescent yeast
assay. Sci Total Environ 378:343–351
146. Furuichi T, Kannan K, Suzuki K, Tanaka S, Giesy JP, Masunaga S (2006) Occurrence of
estrogenic compounds in and removal by a swine farm waste treatment plant. Environ Sci
Technol 40:7896–7902
147. Mahjoub O, Escande A, Rosain D, Casellas C, Gomez E, Fenet H (2011) Estrogen-like and
dioxin-like organic contaminants in reclaimed wastewater: transfer to irrigated soil and
groundwater. Water Sci Technol 63:1657–1662
148. David A, Gomez E, Ait-Aissa S, Rosain D, Casellas C, Fenet H (2010) Impact of urban
wastewater discharges on the sediments of a small Mediterranean river and associated
coastal environment: Assessment of estrogenic and dioxin-like activities. Arch Environ
Contam Toxicol 58:562–575
149. Mnif W, Zidi I, Hassine AIH, Gomez E, Bartegi A, Roig B, Balaguer P (2012) Monitoring
endocrine disrupter compounds in the Tunisian Hamdoun River using in vitro bioassays.
Soil Sediment Contam 21:815–830
150. Maletz S, Floehr T, Beier S, Klumper C, Brouwer A, Behnisch P, Higley E, Giesy JP,
Hecker M, Gebhardt W, Linnemann V, Pinnekamp J, Hollert H (2013) In vitro
characterization of the effectiveness of enhanced sewage treatment processes to eliminate
endocrine activity of hospital effluents. Water Res 47:1545–1557
151. Vethaak AD, Lahr J, Schrap SM, Belfroid AC, Rijs GBJ, Gerritsen A, de Boer J, Bulder AS,
Grinwis GCM, Kuiper RV, Legler J, Murk TAJ, Peijnenburg W, Verhaar HJM, de Voogt P
(2005) An integrated assessment of estrogenic contamination and biological effects in the
aquatic environment of The Netherlands. Chemosphere 59:511–524
152. Houtman CJ, Booij P, van der Valk KM, van Bodegom PM, van den Ende F, Gerritsen
AAM, Lamoree MH, Legler J, Brouwer A (2007) Biomonitoring of estrogenic exposure and
identification of responsible compounds in bream from Dutch surface waters. Environ
Toxicol Chem 26:898–907
153. Leusch FDL, De Jager C, Levi Y, Lim R, Puijker L, Sacher F, Tremblay LA, Wilson VS,
Chapman HF (2010) Comparison of five in vitro bioassays to measure estrogenic activity in
environmental waters. Environ Sci Technol 44:3853–3860
154. Wehmas LC, Cavallin JE, Durhan EJ, Kahl MD, Martinovic D, Mayasich J, Tuominen T,
Villeneuve DL, Ankley GT (2011) Screening complex effluents for estrogenic activity with
the T47D-KBluc cell bioassay: assay optimization and comparison with in vivo responses in
fish. Environ Toxicol Chem 30:439–445
155. Maggioni S, Balaguer P, Chiozzotto C, Benfenati E (2013) Screening of endocrine-
disrupting phenols, herbicides, steroid estrogens, and estrogenicity in drinking water from
the waterworks of 35 Italian cities and from PET-bottled mineral water. Environ Sci Pollut
Res 20:1649–1660
156. Suzuki G, Tue NM, Malarvannan G, Sudaryanto A, Takahashi S, Tanabe S, Sakai S,
Brouwer A, Uramaru N, Kitamura S, Taldgami H (2013) Similarities in the endocrine-
disrupting potencies of indoor dust and flame retardants by using human osteosarcoma
(U2OS) cell-based reporter gene assays. Environ Sci Technol 47:2898–2908
157. Van der Linden SC, Heringa MB, Man HY, Sonneveld E, Puijker LM, Brouwer A, Van der
Burg B (2008) Detection of multiple hormonal activities in wastewater effluents and surface
water, using a panel of steroid receptor CALUX bioassays. Environ Sci Technol
42:5814–5820
158. Blankvoort BMG, Rodenburg RJT, Murk AJ, Koeman JH, Schilt R, Aarts J (2005)
Androgenic activity in surface water samples detected using the AR-LUX assay: indications
for mixture effects. Environ Toxicol Pharmacol 19:263–272
Detection of Organic Compounds with Whole-Cell Bioluminescent Bioassays 151
Abstract Bacteriophages are viruses that exclusively infect bacteria. They are
ideally suited for the development of highly specific diagnostic assay systems.
Bioluminescent reporter bacteriophages are designed and constructed by integra-
tion of a luciferase gene in the virus genome. Relying on the host specificity of the
phage, the system enables rapid, sensitive, and specific detection of bacterial
pathogens. A bioluminescent reporter phage assay is superior to any other
molecular detection method, because gene expression and light emission are
dependent on an active metabolism of the bacterial cell, and only viable cells will
yield a signal. In this chapter we introduce the concept of creating reporter phages,
discuss their advantages and disadvantages, and illustrate the advances made in
developing such systems for different Gram-negative and Gram-positive patho-
gens. The application of bioluminescent reporter phages for the detection of
foodborne pathogens is emphasized.
Abbreviations
d Day(s)
g Gram(s)
h Hour(s)
L Liter(s)
min Minute(s)
ml Milliliter(s)
mol Mole(s)
s Second(s)
Contents
1 Introduction........................................................................................................................ 156
2 Bioluminescence as Reporter System............................................................................... 158
3 Principles of Reporter Bacteriophage Construction and Use .......................................... 159
4 Specific Bioluminescent Reporter Phages........................................................................ 162
4.1 Antibiotic Resistance ................................................................................................ 166
4.2 Combination of Bioluminescent Reporter Phages with Other Technologies......... 166
5 Summary, Conclusions, and Outlook ............................................................................... 167
References................................................................................................................................ 167
1 Introduction
Bacteriophages are viruses that solely infect bacteria and rely on the host
metabolism for reproduction, making them perfect intracellular parasites. They
represent the most abundant biological entities on earth and are a major driving
force for bacterial evolution [1, 2]. Most bacteriophages belong to the order
Caudovirales, the tailed bacteriophages, with the three families Siphoviridae,
Myoviridae, and Podoviridae. Bacteriophages propagate themselves by adsorption
to a host bacteria cell, penetration of the cell wall, and subsequent injection of the
phage DNA or RNA. The phage can follow a temperate lifestyle, which is char-
acterized by the integration of the phage genome into the host chromosome and
coreplication with the bacterial chromosome. In contrast, a lytic lifestyle results in
reprogramming of the host cell by the invading DNA, resulting in production of
new virus progeny and subsequence cell lysis. Temperate phages (in the integrated
state called ‘‘prophage’’) can excise their genome from the host chromosome under
adverse physiological conditions and enter the lytic cycle, thereby eventually
killing the host. In both cases, phage DNA is transduced into the host cell during
the initial infection, a basic step and prerequisite for development of any reporter
bacteriophage.
Bacteriophages have been widely used for therapeutic purposes and for diag-
nostics of pathogenic bacteria for many years. Phage therapy has been one of the
pillars of eastern Europe and Soviet Union healthcare systems but the concept was
completely abandoned in the Western world due to the invention of antibiotics [3,
4]. The current crisis caused by antibiotic-resistant bacteria prompted a reconsid-
eration of bacteriophage therapy, and sparked a new area of research using these
viruses. In addition to application in medical settings, bacteriophages are becoming
increasingly popular for biocontrol of pathogenic bacteria in food. Multiple studies
demonstrate the feasibility of the approach in controlling Salmonella, Campylo-
bacter, Listeria, and many other pathogens in a variety of foods, as well as in
livestock [5–9]. Also, enzymatic components of the phage, such as endolysins, can
be used to control infections and remove unwanted pathogens [10, 11].
Detection of Bacteria with Bioluminescent Reporter Bacteriophage 157
regions is not an easy task. Phage genes are usually densely packed on the gen-
omes, often featuring coding capacities of more than 90 % [31–33]. Moreover,
insertion of large gene fragments into noncoding regions might be difficult to
achieve. Despite the recent advances in sequencing technologies and an increasing
number of phage genomes available, the function of most genes in phage genomes
is not clear [34, 35]. Determining a nonessential gene or a noncoding region in
which a reporter gene may be inserted is therefore not trivial. This fact also
represents the major difficulty in transposition-based methods for generating a
reporter phage: random insertion likely results in disruption of functions required
for infection and multiplication.
A further restriction to the more straightforward design and construction of
reporter phages is that direct cloning is rarely possible. In the best case, chro-
mosomally integrated temperate phages (prophages) will be amenable to genetic
manipulation in the same way as the host chromosome. However, this depends on
the availability of suitable tools for manipulating this organism. In contract,
genetic manipulation of virulent bacteriophage chromosomes is inherently much
more difficult, because the phage DNA is only accessible for modification during
the infection process. The method of choice is homologous recombination and
double cross-over between the mutation locus placed on a plasmid and the phage
DNA, which will result in mutated DNA incorporated into new phage particles.
Because the frequency of successful recombination is very low, the challenge is to
isolate the recombinant virus from the background of wild-type phages. However,
the desired phage can be relatively conveniently selected from the background by
screening for ‘‘bioluminescent plaques’’ formed on infected host cells (in the
presence of the substrate for the luciferase enzyme).
The reporter gene should be placed under control of a strong promoter, active
upon infection of the bacterial host. Naturally, a phage late-gene promoter would
be best suited for this task. Alternatively, nonphage promoters to enhance suffi-
cient levels of reporter gene expression in certain host bacteria could also be used.
The physical structure of the phage genome plays a major role in determining
the correct placement of a reporter gene. Phage genomes must be circularized or
primed by proteins for efficient, loss-free DNA replication in the host cell.
Therefore, most phage genomes feature overlapping single-stranded ends or ter-
minal redundancies to enable circularization upon infection [36], the latter of
which may be extensive [37]. Any genetic manipulation, in particular addition of
sequence, must take into account that phage capsids provide limited space, and
packaging signals must not be destroyed by gene insertion. This is well illustrated
by construction of Listeria reporter phage A511::luxAB. The 2.2 kb luxAB gene
was inserted into the 30 -region of the major capsid protein gene, which features a
strong promoter and is highly expressed during phage replication [38]. At the time
this work was done in 1995, no complete genome sequence and no information
about the A511 genome structure was available. Only 12 years later, the complete
genome sequence of A511 and the 3.125 bp fixed terminal redundancy of its
chromosome were resolved [37]. These findings explained why it was possible to
insert the 2.2 bp luxAB fusion into the A511 genome. However, it is still unclear
Detection of Bacteria with Bioluminescent Reporter Bacteriophage 161
how the A511 terminal redundancy is generated and maintained [37]. In another
example, Kuhn et al. reported severe difficulties in obtaining a transposon-mutated
Felix O1 Salmonella reporter phage, which was attributed to the phage not being
able to accommodate additional genetic information. Eventually, essential genetic
information had to be deleted and supplied in trans to enable recombinant reporter
phage generation [25].
A very important advantage of the detection of bacteria using bioluminescent
phages is the fact that bacteriophages rely on the host cell metabolism for gene
expression and protein synthesis, and the bioluminescence reaction also requires
substrates such as FMNH2 originating from an active metabolism. Consequently,
luciferase and bioluminescence cannot be produced outside a viable host cells. The
reporter phage assay is therefore superior to PCR or any other molecular method,
which cannot distinguish between live and dead cells.
The theoretical detection limit of a reporter phage assay is a single viable cell.
However, this is greatly dependent on the reporter system and detector, and spe-
cific characteristics of the phage, such as its multiplication rate and requirements.
The matrix from which bacteria are isolated, enriched, and detected also plays an
important role. Clearly, there is a significantly better performance of the assay in
broth or buffer than directly in a food matrix [14, 29, 39]. The food surface
provides hiding niches for bacterial target cells, which absorb the majority of
phage particles, or inactivate them by indigenous substances. Also, free diffusion
of the phage particle is essential for successful binding to the target host, a con-
dition not given in most food materials [40]. In most cases, a pre-enrichment step
is strictly required for detection of bacterial cells, with a reasonable lower limit of
10 or 100 cells per ml or g food being detectable [14, 29, 39]. With longer pre-
enrichment, this limit may be lowered down to one cfu/ml/g or less [39], which is
well within the regulatory limits for most foods and pathogens.
Some drawbacks of bioluminescent reporter phage assays should also be
mentioned here, such as the lack of thermostability of the LuxAB fusion from
Vibrio, which is progressively inactivated at temperatures of 30 °C and above,
limiting its use to bacteria that can grow at or below this temperature (the psy-
chotrophic pathogen Listeria represents an ideal case). Also, emission of biolu-
minescence from infected bacteria is transient, and the time window for
measurement is shortly after injection of the aldehyde substrate. This is caused by
the limited supply of FNMH2 in infected cells, which is rapidly depleted by the
bioluminescence reaction, resulting in a short burst of light emission of typically
less than 20 s [15, 38]. Increasing the measurement time will therefore not result in
a lower detection limit, that is, a better signal-to-noise ratio [15].
Furthermore, the use of relatively simple single-tube luminometers is not easily
applicable in large-scale screening of food materials. Bioluminescence measure-
ment from phage-infected bacteria in small liquid samples is much better suited to
multiplates and semiautomated luminescence readers with substrate injection
systems. However, such systems may be relatively expensive, similar to any other
modern high-throughput screening method. Yet, reporter bacteriophages may also
be integrated into lab-on-a-chip approaches. Bacteriophages can be immobilized
162 J. Klumpp and M. J. Loessner
on membranes and other solid supports [19, 41–43], and the detection system
could potentially be miniaturized. Hazbon et al. reported simplification of the
detection of the light-emitting reaction from Mycobacterium reporter phages by
using inexpensive Polaroid film, and achieved equal sensitivity compared to a
luminometer, albeit requiring a longer detection time [20].
Finally, the lack of a suitable bacteriophage for the target organism, or diffi-
culties in genetic manipulation might present major hurdles for development of
other or additional bioluminescent phages.
[25], likely because of the terminally redundant invariable genome ends of this
phage (Klumpp and Marti, unpublished results). The Felix-O1 reporter was
eventually made by replacement of three genes with a luxAB cassette, and in trans
supply of one of the essential genes. However, the reporter phage performed
poorly in strains other than the propagation host [25]. One interesting property of
this specific construct is the fact that it is genetically locked; that is, it can only
reproduce in the complemented laboratory host strain. Therefore, it is unable to
reproduce and spread into the environment, a fact that might help address general
concerns regarding use of GMO reporter phages.
In 1991, Kodikara et al. developed a reporter phage for near-online detection of
enterobacteria. For proof-of-concept, an abattoir environment was used, and meat-
processing surfaces were sampled. The authors reported a 104 cfu/g detection limit
for a 1 h quick test, and could significantly reduce the limit to 10 cfu using a 4 h
sample pre-enrichment [51].
E. coli, namely serotype O157:H7 was also used as a target for reporter phage
detection. The phage was based upon the temperate virus UV10, and featured
promoterless bacterial luxAB genes that were inserted by transposon mutagenesis
on a 3.6-kb fragment [52]. The authors claimed that UV10 was able to detect
O157:H7 after only 1 h of infection, and could detect 64 % of screened E. coli
O157:H7 isolates. Use of a temperate phage is probably the reason for the
insufficient range of detectable isolates, and construction of further reporter phages
was suggested [52].
In 2008, Ripp and coworkers published the construction of an E. coli O157:H7
reporter based on phage PP01 [26]. Earlier, the same phage was engineered as a
fluorescent reporter phage by Oda et al., to present a GFP molecule on its capsid.
The phage enabled specific tagging of target bacteria, which could be seen under
the fluorescence microscope [17]. Ripp et al. engineered the phage to encode the
luxI/R quorum sensing, and a luxCDABE bioluminescence operon. The reporter
phage is described to respond specifically and autonomously to the presence of
E. coli O157:H7. The presence of the LuxI and R proteins serves for signal
amplification. LuxI produces an autoinducer N3–(oxohexanoyl-) L-homoserine
lactone (OHHL), which activates LuxR, which in turn upregulates the lux gene
transcription. The LuxCDE gene products catalyze synthesis of the luciferase
aldehyde substrate, avoiding the need for external substrate addition during
detection [26]. The phage featured a detection limit of 103 cfu/ml from pure
cultures. To push this value down to 1 cfu/ml, an enrichment step was introduced,
resulting in a total assay time of approximately 10–12 h. In apple juice, the
detection limit for E. coli O157:H7 could was 100 cfu/ml, and in water it was
1 cfu/ml. In contrast, the limit was a massive 106 cfu/g from ground beef, possibly
due to a high background bioluminescence because of the intrinsic presence of the
OHHL autoinducer in beef [26]. This points to a major drawback of signal au-
toamplification: the presence of an inducer-like substance can trigger a light-
emitting reaction and might produce false-positive results.
Arguably one of the first reporter phages developed for Gram-positive patho-
gens and successfully applied to foods was Listeria phage A511:luxAB [38].
164 J. Klumpp and M. J. Loessner
Listeria is the causative agent of the rare but severe infection termed listeriosis
[53], and is almost exclusively transmitted via contaminated foods [54, 55].
Culture-based detection methods for Listeria take 4–6 days, which is quite prob-
lematic considering the short shelf-life of most ready-to-eat foods and delicates-
sen. Phage A511 is a virulent myovirus of Listeria and can infect the majority of
Listeria strains [56]. It later became a model organism for the bacteriophage
subfamily Spounavirinae, which comprises many related phages of potential
biotechnological interest [37, 57]. The reporter phage was developed with the goal
to reduce detection time and limits. Current regulations demand the absence of
Listeria from 25 g of food for certain types of food, which is still difficult to
confirm even by the most modern molecular detection methods. In this work, a
luxAB gene cassette from Vibrio harveyi was inserted downstream of the major
capsid gene of A511, and expression is driven by the dedicated and strong Pcps
promoter, resulting in high expression levels. Recombination into the wild-type
phage was achieved by plasmid-based double-crossover during phage infection
and genome replication [38]. Mutant phages could be identified and isolated using
the luciferase activity, and could be calculated to have occurred at a relatively high
frequency of approximately 1:50,000. Maximum signal intensity is approximately
100–140 min postinfection of Listeria target cells [38].
In a follow-up study, the efficacy of A511:luxAB was evaluated in artificially
and naturally contaminated foods [39]. The phage enabled highly sensitive
detection of Listeria contamination in 55 out of 348 tested field samples, with no
false-positives. A pre-enrichment step of 20 h was used to achieve best results,
which is superior to the 4–6 days required for the standard plating method.
Depending on the type of food, detection of 0.1 cell/g could be achieved, whereas
in other, more complex types of food, such as minced meat, positive diagnostics of
10 cells/g food were possible [39]. It should be noted that A511 is not species
specific, and can infect and yield signals with other Listeria species. This is not
necessarily a drawback, inasmuch as any Listeria contamination is considered
undesirable and a marker of poor hygiene or contaminated raw products [58].
More recently, the same concept was used to modify A511 with a different
reporter system. The celB glycosidase from Pyrococcus furiosus features extreme
heat stability—in contrast to the original luxAB system, which is instable above
35 °C—and sufficient pH stability. The enzyme is most active at 102–106 °C and
pH 5–5.5, a major advantage compared to the temperature-sensitive luciferase
enzyme [15]. Because of its versatile activity, featuring both b-galactosidase and
b-glycosidase activity, a wide range of chromogenic, fluorescent, or chemilumi-
nescent substrates can be used for pathogen detection. The system has been proven
to be suitable for detection of Listeria from the food matrix, with detection limits
as low as 10 cells. The assay can be fully automated to 96-well plate format [15].
One advantage of the celB reporter phage is that detection does not need to be
carried out in a specific time window as with luciferase activity, but the color
conversion of a cromogenic substrate is an end point assay and can be determined
any time after incubation.
Detection of Bacteria with Bioluminescent Reporter Bacteriophage 165
The same authors also produced a reporter phage for Yersinia pestis [67], which
(in an improved version) could potentially also be used to assess antibiotic sus-
ceptibility of this pathogen. Yersinia typing phage UA1122 was equipped with the
bacterial luxAB genes in a noncoding region, and the ability of the phage to
generate a drug-concentration-dependent signal was harnessed [68].
Plant pathogenic bacteria also represent targets for the development of reporter
bacteriophages, as they cause significant losses in agriculture, and early-warning
and screening systems are needed. Schofield et al. report the development of phage
PBSPCA1::luxAB, which specifically targets Pseudomonas cannabina pv. alisal-
ensis, the causative agent of bacterial blight of crucifers [28]. The Vibrio harveyi
luxAB genes were inserted into an phoH homologue in the phage genome, under
control of a strong bacterial promoter [28]. The detection limit was 260 cfu/ml, at
approximately 120 min after infection [28, 69].
The detection limit of bioluminescent reporter phages can be further and signifi-
cantly improved if immunomagnetic separation technologies are used in pre-
enrichment of target bacteria. Especially suitable seems the use of cell-wall
binding domains of bacteriophage endolysins coated on paramagnetic beads for
Detection of Bacteria with Bioluminescent Reporter Bacteriophage 167
fast and highly efficient enrichment of pathogens [71, 72]. Pre-enriching the target
bacteria with magnetic separation, followed by detection with bioluminescent
reporter phages could lower the detection limit by several orders of magnitude, and
provide ultrafast diagnostics of viable bacteria only (Kretzer and Loessner
unpublished data). The methods may be automated and performed in a high-
throughput setting. Favrin et al. used classical immunomagnetic separation for this
purpose, and were able to detect as low as 3 cfu Salmonella enteritidis per 25 g of
food [73].
References
43. Arya SK, Singh A, Naidoo R, Wu P, McDermott MT, Evoy S (2011) Chemically
immobilized T4-bacteriophage for specific Escherichia coli detection using surface plasmon
resonance. The Analyst 136:486–492
44. Castilho BA, Olfson P, Casadaban MJ (1984) Plasmid insertion mutagenesis and lac gene
fusion with mini-mu bacteriophage transposons. J Bacteriol 158:488–495
45. Ulitzur S, Kuhn J (1989) Detection and/or identification of microorganisms i a test sample
using bioluminescence or other exogenous genetically introduced marker, Patent C12N15/52,
06/739,957 USPTO
46. Kuhn J, Suissa M, Chiswell D, Azriel A, Berman B, Shahar D, Reznick S, Sharf R, Wyse J,
Bar-On T, Cohen I, Giles R, Weiser I, Lubinsky-Mink S, Ulitzur S (2002) A bacteriophage
reagent for Salmonella: molecular studies on Felix 01. Int J Food Microbiol 74:217–227
47. Chen J, Griffiths M (1996) Salmonella detection in egg using Lux + bacteriophages. J Food
Prot 59:908–914
48. Stewart G, Smith T, Denyer S (1989) Genetic engineering for bioluminescent bacteria. Food
Sci Technol Today 3: 19-22
49. Turpin P, Maycroft KA, Bedford J, Rowlands CL (1993) A rapid luminescent-phage based
MPN method for the enumeration of Salmonella typhimurium in environmental samples. Lett
Appl Microbiol 16: 24-27
50. Thouand G, Vachon P, Liu S, Dayre M, Griffiths MW (2008) Optimization and validation of
a simple method using P22:luxAB bacteriophage for rapid detection of Salmonella enterica
serotypes A, B, and D in poultry samples. J Food Prot 71:380–385
51. Kodikara CP, Crew HH, Stewart GS (1991) Near on-line detection of enteric bacteria using
lux recombinant bacteriophage. FEMS Microbiol Lett 83:261–266
52. Waddell TE, Poppe C (2000) Construction of mini-Tn10luxABcam/Ptac-ATS and its use for
developing a bacteriophage that transduces bioluminescence to Escherichia coli O157:H7.
FEMS Microbiol Lett 182:285–289
53. Vazquez-Boland JA, Kuhn M, Berche P, Chakraborty T, Dominguez-Bernal G, Goebel W,
Gonzalez-Zorn B, Wehland J, Kreft J (2001) Listeria pathogenesis and molecular virulence
determinants. Clin Microbiol Rev 14:584–640
54. Farber JM, Peterkin PI (1991) Listeria monocytogenes, a food-borne pathogen. Microbiol
Rev 55:476–511
55. McLauchlin J, Mitchell RT, Smerdon WJ, Jewell K (2004) Listeria monocytogenes and
listeriosis: a review of hazard characterisation for use in microbiological risk assessment of
foods. Int J Food Microbiol 92:15–33
56. Loessner MJ, Busse M (1990) Bacteriophage typing of Listeria species. Appl Environ
Microbiol 56:1912–1918
57. Klumpp J, Lavigne R, Loessner MJ, Ackermann HW (2010) The SPO1-related
bacteriophages. Arch Virol 155:1547–1561
58. Hagens S, Loessner MJ (2007) Luciferase Reporter Bacteriophages. In: Marks RS, Cullen
DC, Karube I, Lowe CR, Weetall HH (eds) Handbook of Biosensors and Biochips. Wiley,
Hoboken
59. Pearson RE, Jurgensen S, Sarkis GJ, Hatfull GF, Jacobs WR Jr (1996) Construction of D29
shuttle phasmids and luciferase reporter phages for detection of mycobacteria. Gene
183:129–136
60. Jacobs WR Jr, Barletta RG, Udani R, Chan J, Kalkut G, Sosne G, Kieser T, Sarkis GJ, Hatfull
GF, Bloom BR (1993) Rapid assessment of drug susceptibilities of Mycobacterium
tuberculosis by means of luciferase reporter phages. Science 260:819–822
61. Piuri M, Jacobs WR Jr, Hatfull GF (2009) Fluoromycobacteriophages for rapid, specific, and
sensitive antibiotic susceptibility testing of Mycobacterium tuberculosis. PLoS ONE 4:e4870
62. Banaiee N, Bobadilla-Del-Valle M, Bardarov S Jr, Riska PF, Small PM, Ponce-De-Leon A,
Jacobs WR Jr, Hatfull GF, Sifuentes-Osornio J (2001) Luciferase reporter
mycobacteriophages for detection, identification, and antibiotic susceptibility testing of
Mycobacterium tuberculosis in Mexico. J Clin Microbiol 39:3883–3888
Detection of Bacteria with Bioluminescent Reporter Bacteriophage 171
63. Banaiee N, Bobadilla-del-Valle M, Riska PF, Bardarov S Jr, Small PM, Ponce-de-Leon A,
Jacobs WR Jr, Hatfull GF, Sifuentes-Osornio J (2003) Rapid identification and susceptibility
testing of Mycobacterium tuberculosis from MGIT cultures with luciferase reporter
mycobacteriophages. J Med Microbiol 52:557–561
64. Rondon L, Piuri M, Jacobs WR Jr, de Waard J, Hatfull GF, Takiff HE (2011) Evaluation of
fluoromycobacteriophages for detecting drug resistance in Mycobacterium tuberculosis.
J Clin Microbiol 49:1838–1842
65. Schofield DA, Molineux IJ, Westwater C (2011) ‘Bioluminescent’ reporter phage for the
detection of category A bacterial pathogens. J Vis Exp 53:e2740
66. Schofield DA, Westwater C (2009) Phage-mediated bioluminescent detection of Bacillus
anthracis. J Appl Microbiol 107:1468–1478
67. Schofield DA, Molineux IJ, Westwater C (2009) Diagnostic bioluminescent phage for
detection of Yersinia pestis. J Clin Microbiol 47:3887–3894
68. Schofield DA, Molineux IJ, Westwater C (2012) Rapid identification and antibiotic
susceptibility testing of Yersinia pestis using bioluminescent reporter phage. J Microbiol
Methods 90:80–82
69. Schofield D, Bull CT, Rubio I, Wechter WP, Westwater C, Molineux IJ (2013) ‘‘Light-
tagged’’ bacteriophage as a diagnostic tool for the detection of phytopathogens.
Bioengineered 4:50–54
70. Carriere C, Riska PF, Zimhony O, Kriakov J, Bardarov S, Burns J, Chan J, Jacobs WR Jr
(1997) Conditionally replicating luciferase reporter phages: improved sensitivity for rapid
detection and assessment of drug susceptibility of Mycobacterium tuberculosis. J Clin
Microbiol 35:3232–3239
71. Schmelcher M, Shabarova T, Eugster MR, Eichenseher F, Tchang VS, Banz M, Loessner MJ
(2010) Rapid multiplex detection and differentiation of Listeria cells by use of fluorescent
phage endolysin cell wall binding domains. Appl Environ Microbiol 76:5745–5756
72. Kretzer JW, Lehmann R, Schmelcher M, Banz M, Kim KP, Korn C, Loessner MJ (2007) Use
of high-affinity cell wall-binding domains of bacteriophage endolysins for immobilization
and separation of bacterial cells. Appl Environ Microbiol 73:1992–2000
73. Favrin SJ, Jassim SA, Griffiths MW (2003) Application of a novel immunomagnetic
separation-bacteriophage assay for the detection of Salmonella enteritidis and Escherichia
coli O157:H7 in food. Int J Food Microbiol 85:63–71
74. Marti R, Zurfluh K, Hagens S, Pianezzi J, Klumpp J, Loessner MJ (2013) Long tail fibers of
the novel broad host range T-even bacteriophage S16 specifically recognize Salmonella
OmpC. Mol Microbiol 87:818–834
Part IV
Applications of Bioluminescence
in Health
Application of Enzyme Bioluminescence
for Medical Diagnostics
Keywords Bioluminescence
Ca2+-regulated photoprotein Diagnostics
Immunoassay Luciferase Nucleic acid hybridization assay
Abbreviations
Ab Antibody
Ag Antigen
ATP Adenosine-50 -triphosphate
BRET Bioluminescence resonance energy transfer
CMV Cytomegalovirus
ELISA Enzyme-linked immunosorbent assay
EYFP Yellow fluorescent protein
GFP Green fluorescent protein
hCG Human chorionic gonadotropin
Contents
1 Introduction........................................................................................................................ 176
2 Immunoassay: State-of-the-Art ......................................................................................... 177
2.1 Luciferases as Labels for Immunoassay .................................................................. 179
3 Nucleic Acid Hybridization Assay Based on Bioluminescent Reporters ....................... 183
3.1 Bioluminescent Assay of Infectious Agents............................................................ 185
3.2 Bioluminescent Assay in Oncology Diagnostics..................................................... 187
4 Multianalytical Bioluminescent Assay ............................................................................. 189
5 Bioluminescent Binding Assay in Homogeneous Format ............................................... 192
6 Summary, Conclusions, and Outlook ............................................................................... 193
References................................................................................................................................ 194
1 Introduction
At present, among bioluminescent systems the best-known are those derived from
fireflies, marine bacteria, coelenterates, and copepods. ‘‘Best-known’’ here implies
availability of cDNAs coding corresponding light-emitting proteins and affordable
Application of Enzyme Bioluminescence for Medical Diagnostics 177
2 Immunoassay: State-of-the-Art
Fig. 1 Scheme for: a sandwich-type and b competitive immunoassays, and c basic antibody
structure (human IgG) demonstrating pairs of identical heavy (blue) and light (yellow) chains.
Both pairs contain variable domens, VH (dark blue) and VL (dark yellow), responsible for antigen
binding
factors: the efficiency of the immunocomplex formation and the ability of the
detection system to register this complex with high sensitivity. Complex formation
is provided by antibodies’ specificity and binding affinity to the antigen. Anti-
bodies are proteins, which are produced in animals by immunological response to
the presence of a foreign substance (Fig. 1c schematically presents a basic anti-
body structure). Antibodies are a crucial component of immunoassay performance
due to their ability to bind to an extremely wide range of natural and man-made
molecules, cells, and viruses; exceptional specificity for the analyte that enables us
to assay it in complex biological media (sera, urine, etc.); and the strength of
binding, providing the formation of a strong noncovalent complex Ag–Ab that
survives at processing and signal generation.
The immunocomplex detection system (signal generation plus measuring tool)
accounts for immunoassay sensitivity. The prime requirement for the one is the
detection of the label above background noise. A wide range of labels and their
nature-dependent detection systems are applied in immunological assay.
Radiolabels are the first to be used but the problems dealing with radioactive wastes
and inherent instability of radiolabeled reagents have stimulated the development
Application of Enzyme Bioluminescence for Medical Diagnostics 179
was developed using the luciferase genetically fused with antigene or antibody.
Figure 3 displays the luciferase immunoprecipitation system (LIPS), developed by
Burbelo and coauthors to detect antibodies responsible for tumor-associated
proteins [10], and a variety of infectious agents [11–13]. Briefly, this approach
involves: (a) construction of antigen–RLuc fusion and expression of the fusion
protein in mammalian COS cells; (b) incubation of RLuc–antigen fusion and
analyzed sera sample to bind antibody of interest; (c) immobilization of the
complex RLuc–antigen fusion–antibody on protein A/G beads, and (d) quantita-
tion of antigen-specific antibody by adding coelenterazine and measuring light
production. The most important point of this technology is the mammalian
production of fusion protein to provide correct folding and specific posttransla-
tional modifications of the antigenic domain, which are impossible under bacterial
expression. Of course it makes the analysis more expensive, although in some
crucial cases it is justifiable by high sensitivity.
Fusion proteins RLuc–antibody have mostly been engineered for ex vivo or
in vivo analytical applications. For these constructions, the variable regions
VL and VH chains of immunoglobulin (Fig. 1c) being minimal engineered anti-
body fragments, capable of efficient target binding, are usually used as luciferase
partners. Under bacterial expression, periplasmic secretion of chimeric protein is
usually employed in order to provide antibody domain affinity. As shown for
anticarcinoembryonic antigen antibody–RLuc fusion proteins [14], bacterial
expression allowed purification of small amounts of proteins, with only about half
of them recovered intact, whereas under mammalian expression the recovery was
greater, and owing to the lack of proteases, the protein purified remained stable for
several months when stored at 4 °C.
Luciferases from M. longa and G. princeps are single-chain polypeptides
(20–24 kDa) that contain 10 Cys residues, organizing several disulfide bonds. The
bacterial expression gives mostly unfolded proteins and the obtaining of an active
protein requires a special refolding procedure or expression in eukaryotic cells.
182 L. A. Frank and V. V. Krasitskaya
tolerance to chemical modifications, and conjugation with the other molecules. The
conjugates in solution as well as in frozen and lyophilized states were found to be
stable and well-stored. A great number of publications described photoprotein-
based immunoassay of different analytes of clinical interest: hormones, interleu-
kins, oncomarkers, and the like, and infections (see [24]). The assays were carried
out in different formats (sandwich, competitive), and performed in solid-phase,
homogeneous, and even in flow injection variants. The papers consider investiga-
tions of model samples as well as sera, saliva, and mucous of patients and exper-
imental animals. Clinical trials began in 1994. At that time aequorin-based
thyrotropin (TSH) assay (developed and marketed by SeaLite Inc., USA) was tested
in the Pathology and Laboratory Medicine Departments of Emory University and
Veterans Administration Hospital (Atlanta, USA) [25]. The authors measured
serum TSH in 153 euthyroid individuals with thyroidal illnesses (primary hypo- and
hyperthyroids, thyroid cancer, etc.) applying bioluminescent methods and com-
pared the results with the data obtained by commercially available Nichols, ACS-
180 and TOSOH methods. The aequorin-based method was found to have the
required performance characteristics (e.g., the functional sensitivity of 0.017 mIU/
L, which is higher than in the cases of ACS-180 and TOSOH assays) and is ‘‘clearly
qualified as a last, third-generation TSH assay.’’
From the above presented data it appears that the application of classical
luciferases as reporters in immunoassay is complicated by their instability to
chemical conjugation with biospecific molecules (recognition elements) or by the
problems of genetically fused proteins’ folding under their bacterial expression. By
contrast, Ca2+-regulated photoproteins are free of these shortcomings and this
makes them perfect in vitro reporting molecules. The immunoassay on their basis
is fast and simple and, what is most important, highly sensitive in detecting the
target.
But more often, quantitative PCR analysis is important for evaluation of therapy
effectiveness, detection of gene expression through reverse transcriptase-poly-
merase chain reaction (RT-PCR), and so on.
Many strategies have been developed to quantitate PCR products. The tradi-
tional approach of calculation is visualization by DNA band density after sepa-
ration on agarose gel, or scintillation counting of radiolabeled products. The
obvious shortcomings of these techniques—low sensitivity, bulkiness to perform,
or necessity to use radioactive materials—hamper implementation of these assays
in routine settings. The assay improvement was achieved by using automated
capillary electrophoresis and laser-induced fluorescence [26]. The need for
electrophoresis was avoided by using magnetic beads coated with streptavidin to
capture biotinylated PCR fragments [27]. After hybridization with a hapten-
labeled probe, these beads were analyzed either by flow cytometry or by immu-
noenzymatic assay based on chemiluminescent, fluorescent, or colorimetric labels.
Quantitative realtime PCR assay allows continuous monitoring of accumulation of
PCR products during the amplification reaction. This provides identification of the
cycle of near-logarithmic PCR product generation (threshold cycle) and, by
inference, the relative quantification of the template DNA present at the start of the
reaction. Because the amplification products are monitored in realtime as being
formed cycle by cycle, no postamplification handling is required. The absolute
quantification is performed according to either an internal standard coamplified
with the sample DNA, or to an external standard curve obtained by parallel
amplification of serial known concentrations of a reference DNA sequence.
However, realtime PCR has certain disadvantages such as the high cost of the
equipment and requirements for high technical skills. Despite the use of high
temperatures, nonspecific primer annealing may occur during PCR, and errors will
be exponentially amplified through multiple cycles. In order to discriminate these
products and to obtain a higher level of specificity, a hybridization step should be
included in assays.
Several methods have been developed for quantitating PCR products using a
microplate format. The tag can be attached to the DNA during the PCR reaction or
via a labeled probe. The PCR product is immobilized on a surface using biotin–
streptavidin, digoxigenin–antidigoxigenin, direct chemical conjugation, or other
methods. The DNA is then detected with a colorimetric, fluorescent, or lumines-
cent tag. The obvious advantages of microplate assays are low cost, fewer steps,
and the possibility to handle a large number of samples simultaneously. However,
in the desirable exponential PCR reaction phase, the concentration of a product is
often too low to be detected by standard methods. This technical hurdle may be
overcome using sensitive detection methods.
In recent years, bioluminescence has found rapidly expanding application in
DNA/RNA assays. With no excitation required, bioluminometric assay offers
higher detectability, wider linear range, and much simpler instrumentation than
fluorometric methods. A variety of hybridization assays has been published, most
of them describing the use of Ca2+-regulated photoproteins as reporters. The first
reports describing the use of recombinant aequorin as a reporter molecule in a
Application of Enzyme Bioluminescence for Medical Diagnostics 185
Fig. 4 Bioluminometric
nucleic acid hybridization
assays with recombinant
aequorin as a reporter
proposed by a Xiao [28] and
Siddigi [30], and b Galvan
[29]. D, digoxigenin, Aeq,
aequorin
The need for detection of minority mutation (i.e., a few mutants within a high
excess of wild-type alleles) arises frequently in the field of cancer genetics.
Routine tumor biopsies often consist of an inhomogeneous mixture of stromal cells
plus tumor cells encompassing a wide range of genetic profiles and mutations.
Quantification of the mutant allele burden (percentage of the mutant allele) is
critical for diagnosis, monitoring of therapy, and detection of minimal residual
disease. Detection of minority mutation is often important in mitochondrial disease
and mutation/polymorphism screening of pooled DNA from many individuals.
With point mutations, the challenge is to quantify the mutant allele while dis-
criminating from a large excess of the normal allele that differs in a single base-
pair. Tsiakalou et al. reported the first bioluminometric assay for quantification of
the allele burden and its application to JAK2 V617F somatic point mutation [39].
This mutation is known to be strongly associated with the myeloproliferative
disorders polycythemia vera and essential thrombocythemia [40]. The proposed
method is performed in microtiter wells and involves a single PCR for amplifi-
cation of both alleles, followed by primer extension (PEXT) reactions with allele-
specific primers (three cycles). The products are captured in microtiter wells and
detected by oligo(dT)-conjugated photoprotein aequorin. The authors
demonstrated that the luminescence signal from the mutant allele is linearly related
to the allele burden. As low as 0.85 % of the mutant allele can be detected and the
linearity extends up to 100 %. The assay is complete within 50 min after the
amplification step. The method can be applied to a large number of reported
somatic mutations where quantification of the allele burden is required.
Iliadi et al. developed two bioluminescent methods that enable absolute
quantification of the allele, with this mutation as an example [41]. The first method
exploits the ability of a nonextendable locked nucleic acid (LNA) effectively to
inhibit the PCR-amplification of the normal allele while the amplification of the
mutant allele remains unaffected. The second method employs allele-specific PCR
primers, thereby allowing the amplification of the corresponding allele only. In
both assays, absolute quantification of the mutant allele is achieved through
coamplification of the recombinant DNA internal standard (DNA competitor). The
amplified products from the target and internal standard are quantified by a
hybridization assay performed at microtiter wells and exploiting the advantages of
Application of Enzyme Bioluminescence for Medical Diagnostics 189
the aequorin reporter. The ratio of luminescence values for target DNA and DNA
competitor linearly relate to the number of JAK2 V617F allele copies initially
present in the sample. The methods allow absolute quantification of less than 300
copies of the mutant allele even in samples containing less than 1 % of the mutant
allele. The novelty and advantages of the proposed method are: (a) the concept of
competitive PCR is exploited for the absolute quantification of the mutant and
normal alleles in single-point mutations using properly designed recombinant
DNA internal standards; (b) the combination of the above principle with a
high-throughput and highly sensitive bioluminometric assay; (c) the considerably
lower cost of instrumentation and reagents compared to other methods, such as
realtime PCR and BEAMing technology; and (d) incorporation of LNA probes in
the competitive PCR simplifies the methods because it does not require two
allele-specific primers.
Bioluminescence-based systems for detection of nucleic acids appeared to be a
powerful and flexible analytical tool for accurate and rapid assay. In the case of
photoproteins, sufficient assay sensitivity allows the detection of amplified product
before the linear relationship of the target to product is lost, as well as the direct
detection of low copies of unamplified target. The use of such analytical systems
considerably improves the assays in terms of simplicity and costs.
Fig. 6 Dual analyte single-well bioluminescence assay based on photoprotein obelin variants
with substantially altered bioluminescence spectra and kinetics. a Bioluminescence of mixture of
obelins transmitted through filter I (fast violet signal) and filter II (slow green signal); dashed
line, time for filter replacement. Upper inset, bioluminescence spectra of obelins (colored lines)
and optical filter transmission (black lines). r.l.u., relative light units. b Scheme for simultaneous
solid-phase immunoassay of two targets
Fig. 7 Principle of
bioluminometric SNP
genotyping by
oligonucleotide ligation
reaction. N probe is specific
for normal allele, M probe for
mutant allele, and C probe is a
common probe. BSA, bovine
serum albumin; ALP, alkaline
phosphatase
measured first by adding Ca2+, then the wells were washed and the chemilumi-
nescence of bound alkaline phosphatase was measured after incubation with
substrate. The ratio of the obtained luminescence signals gives the genotype of
each sample.
The same dual-analyte bio/chemiluminometric assay but for detection of PEXT
reaction products was applied by Konstantou et al. for two SNPs of human
mannose-binding lectin (MBL2) gene (-550 and -221) and one SNP of
cytochrome P450 gene CYP2D6 (CYP2D6*3) [48]. MBL2 is a key component of
the innate immune system, and its deficiency is associated with the increased
susceptibility to various infections and autoimmune disorders. The CYP2D6 is
important because it is involved in the metabolism of many commonly prescribed
drugs [49]. PCR-amplified DNA fragments that span the SNP of interest are
subjected to two PEXT reactions using normal and mutant primers in the presence
of digoxigenin-dUTP and biotin-dUTP. The primers perfectly complementary with
the target DNA are extended by DNA polymerase thus forming digoxigenin-
labeled or biotin-labeled products that are then mixed and analyzed by dual bio/
chemiluminometric assay. The proposed method provides good discrimination
between the two alleles. Patient genotypes showed 100 % concordance with direct
DNA sequencing data.
Elenis et al. developed a method of simultaneous genotyping of two common
SNPs within the toll-like receptor 4 (TLR4) gene, that is, A896G and C1196T [50].
The method consists of a single PCR of the region spanning both polymorphic
sites, followed by a quadruple PEXT reaction in a single tube. Biotinylated
nucleotide is incorporated into extended primers. All four products are captured on
streptavidin-coated microtiter wells and detected with a combination of four
reporters, aequorin, and alkaline phosphatase, b-galactosidase, and horseradish
peroxidase. Bio/chemiluminescence of each reporter was measured sequentially
after washing the wells and incubation with a corresponding substrate. For each
SNP, 46 individuals were genotyped. The accuracy of this method was confirmed
192 L. A. Frank and V. V. Krasitskaya
signals for different buffers and selected pHs, and is insensitive to complicated
sample matrices such as serum. The pair has a large luminescence spectra separation
(110 nm) and provides high assay sensitivity. Instead of GFP the quantum dots are
used as energy acceptors in nucleic acid hybridization assays [56, 57] or synthetic
fluorochromes such as Cy3 or Cy3.5 [58].
In split-protein analytical systems, two polypeptides serve as biospecific rec-
ognition elements attached to the fragmented reporter. The protein domains
interact with the desired target resulting in a ternary complex that drives the
reassembly of the split-protein reporter recovering its activity [59] (Fig. 8b). For
successful creation of a split reporter, a few criteria must be met: each reporter
fragment by itself should not exhibit any activity, the affinity of fragments in the
absence of the target should be negligible, and the reassembled split-protein must
provide an easily measurable readout. This strategy has been successfully used in
reassembling several luciferases as reporters, from R. reniformis [60], firefly [61],
G. princeps [62], and photoprotein aequorin [63]. Here are only several examples
but in reality, the range of biological activities and processes that can be monitored
with a bioluminescence readout using split-protein reassembly both in in vitro and
in vivo settings is unprecedented.
References
8. Stepanyuk GA, Unch J, Malikova NP, Markova SV, John Lee J, Vysotski ES (2010)
Coelenterazine-v ligated to Ca2+-triggered coelenterazine-binding protein is a stable and
efficient substrate of the red-shifted mutant of Renilla muelleri luciferase. Anal Bioanal Chem
398:1809–1817
9. Krasitskaya VV, Burakova LP, Pyshnaya IA, Frank LA (2012) Bioluminescent reporters for
identification of gene allelic variants. Rus J Bioorg Chem 38(3):298–305
10. Burbelo PD, Goldman R, Mattson TL (2005) A simplified immunoprecipitation method for
quantitatively measuring antibody responses in clinical sera samples by using mammalian-
produced Renilla luciferase-antigen fusion proteins. BMC Biotechnol 5(22):692–699
11. Burbelo PD, Ching KH, Mattson TL et al (2007) Rapid antibody quantification and
generation of whole proteome antibody response profiles using LIPS (luciferase
immunoprecipitation systems). Biochem Biophys Res Commun 352:889–895
12. Ramanathan R, Burbelo P, Groot S et al (2008) A luciferase immunoprecipitation systems
assay enhances the sensitivity and specificity of diagnosis of Strongyloides stercoralis
infection. J Infect Dis 198:444–451
13. Burbelo PD, Issa AT, Ching KH, Cohen JI, Iadarola MJ, Marques A (2010) Rapid, simple,
quantitative and highly sensitive antibody detection for lyme disease. Clin Vaccine Immunol
17(6):904–909
14. Venisnik KM, Olafsen T, Loening AM, Iyer M, Gambhir SS, Wu AM (2006) Bifunctional
antibody-Renilla luciferase fusion protein for in vivo optical detection of tumors. Protein Eng
Des Sel 19(10):453–460
15. Markova SV, Golz S, Frank LA, Kalthof B, Vysotski ES (2004) Cloning and expression of
cDNA for a luciferase from the marine copepod Metridia longa. J Biol Chem
279(5):3212–3217
16. Borisova VV, Frank LA, Markova SV, Burakova LP, Vysotski ES (2008) Recombinant
Metridia luciferase isoforms: expression, refolding and applicability for in vitro assay.
Photochem Photobiol Sci 7:1025–1031
17. Verhaegen M, Christopoulos TK (2002) Overexpression, purification and analytical
application of a bioluminescent reporter for DNA hybridization. Anal Chem 74:4378–4385
18. Nakajima Y, Kobayashi K, Yamagishi K, Enomoto T, Ohmiya Y (2004) cDNA cloning and
characterization of a secreted luciferase from the luminous japanese ostracod, Cypridina
noctiluca. Biosci Biotechnol Biochem 68:565–570
19. Wu C, Kawasaki K, Ogawa Y, Yoshida Y, Ohgiya S, Ohmiya Y (2007) Preparation of
biotinylated Cypridina luciferase and its use in bioluminescent enzyme immunoassay. Anal
Chem 79:1634–1638
20. Wu C, Irie S, Yamamoto S, Ohmiya Y (2009) A bioluminescent enzyme immunoassay for
prostaglandin E2 using Cypridina luciferase. Luminescence 24:131–133
21. Liu ZJ, Vysotski ES, Chen CJ, Rose JP, Lee J, Wang BC (2000) Structure of the Ca2+-
regulated photoprotein obelin at 1.7 Å resolution determined directly from its sulfur
substructure. Protein Sci 9:2085–2093
22. Head JF, Inouy S, Teranishi K, Shimomura O (2000) The crystal structure of the photoprotein
aequorin at 2.3 Å resolution. Nature 405:372–376
23. Markova SV, Vysotski ES, Blinks JR, Burakova LP, Wang BC, Lee J (2002) Obelin from the
bioluminescent marine hydroid Obelia geniculata: cloning, expression, and comparison of
some properties with those of other Ca2+-regulated photoproteins. Biochemistry
41:2227–2236
24. Frank LA (2010) Ca2+-regulated photoproteins: effective immunoassay reporters. Sensors
10:11287–11300
25. Sgoutas DS, Tuten TE, Verras AA, Love A, Barton EG (1995) AquaLite bioluminescence
assay of thyrotropin in serum evaluated. Clin Chem 41:1637–1643
26. Fasco MJ, Treanor CP, Spivack S, Figge HL, Kaminsky LS (1995) Quantitative RNA-
polymerase chain reaction-DNA analysis by capillary electrophoresis and laser-induced
fluorescence. Anal Biochem 224:140–147
196 L. A. Frank and V. V. Krasitskaya
27. Vlieger AM, Medenblik AM, van Gijlswijk RP, Tanke HJ, van der Ploeg M, Gratama JW,
Raap AK (1992) Quantitation of polymerase chain reaction products by hybridization-based
assays with fluorescent, colorimetric, or chemiluminescent detection. Anal Biochem 205:1–7
28. Xiao L, Chumfu Y, Nelson CO (1996) Quantitation of RT-PCR amplified cytokine mRNA by
aequorin-based bioluminescence immunoassay. J Immunol Methods 199:139–147
29. Galvan B, Christopoulos TK (1996) Bioluminescence hybridization assay using recombinant
aequorin. Application to the detection of prostate-specific antigen mRNA. Anal Chem
68:3545–3550
30. Siddigi AM, Jennings VM, Kidd MR, Actor JK, Hunter RL (1996) Evaluation of
electrochemiluminescence and bioluminescence-based assays for quantitating specific
DNA. J Clin Lab Anal 10:423–431
31. Actor JK (2000) Bioluminescent quantitation and detection of gene expression during
infectious disease. Comb Chem High Throughput Screen 3(4):277–288
32. Guenthner PC, Hart CE (1998) Quantitative, competitive PCR assay for HIV-1 using a
microplate-based detection system. Biotechniques 24(5):810–816
33. Song X, Coombes BK, Mahony JB (2000) Quantitation of Chlamidia trachomatis 16S rRNA
using NASBA amplification and bioluminescent microtiter plate assay. Comb Chem High
Throughput Screen 3(4):303–313
34. Coombes BK, Mahony JB (2000) Nucleic acid sequence based amplification (NASBA) of
Chlamydia pneumoniae major outer membrane protein (ompA) mRNA with bioluminescent
detection. Comb Chem High Throughput Screen 3(4):315–327
35. Doleman L, Davies L, Rowe L, Moschou EA, Deo S, Daunert S (2007) Bioluminescence
DNA hybridization assay for Plasmodium falciparum based on the photoprotein aequorin.
Anal Chem 79:4149–4153
36. Cissell KA, Rahimi Y, Shrestha S (2008) Bioluminescence-based detection of microRNA,
miR21 in breast cancer cells. Anal Chem 80:2319–2325
37. Zhu S, Si ML, Wu H, Mo YY (2007) MicroRNA-21 targets the tumor suppressor gene
tropomyosin 1 (TPM1). J Biol Chem 282:14328–14336
38. Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY (2007) miR-21-mediated tumor growth. Oncogene
26:2799–2803
39. Tsiakalou V, Petropoulou M, Ioannou PC (2009) Bioluminometric assay for relative
quantification of mutant allele burden: application to the oncogenic somatic point mutation
JAK2 V617F. Anal Chem 81:8596–8602
40. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, Boggon TJ, Wlodarska I,
Clark JJ, Moore S, Adelsperger J, Koo S, Lee JC, Gabriel S, Mercher T, D’Andrea A,
Fröhling S, Döhner K, Marynen P, Vandenberghe P, Mesa RA, Tefferi A, Griffin JD, Eck MJ,
Sellers WR, Meyerson M, Golub TR, Lee SJ, Gilliland DG (2005) Activating mutation in the
tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid
metaplasia with myelofibrosis. Cancer Cell 7(4):387–397
41. Iliadi A, Petropoulou M, Ioannou PC, Christopoulos TK, Anagnostopoulos NI, Kanavakis E,
Traeger-Synodinos J (2011) Absolute quantification of the alleles in somatic point mutations
by bioluminometric methods based on competitive polymerase chain reaction in the presence
of a locked nucleic acid blocker or an allele-specific primer. Anal Chem 83:6545–6551
42. Ito K, Nishimura W, Maeda M, Gomi K, Inouye S, Arakawa H (2007) Highly sensitive and
rapid tandem bioluminescent immunoassay using aequorin labeled Fab fragment and
biotinylated firefly luciferase. Anal Chem Acta 58:245–251
43. Ohkuma H, Abe K, Kosaka Y, Maeda M (2000) Detection of luciferase having two kinds of
luminescent colour based on optical filter procedure: application to an enzyme immunoassay.
Luminescence 15:21–27
44. Frank LA, Borisova VV, Markova SV, Malikova NP, Stepanyuk GA, Vysotski ES (2008)
Violet and greenish photoprotein obelin mutants for reporter applications in dual-color assay.
Anal Bioanal Chem 391:2891–2896
Application of Enzyme Bioluminescence for Medical Diagnostics 197
45. Kudryavtsev AN, Krasitskaya VV, Petunin AI, Burakov AY, Frank LA (2012) Simultaneous
bioluminescent immunoassay of serum total and IgG-bound prolactins. Anal Chem
84:3119–3124
46. Krasitskaya VV, Kudryavtsev AN, Shimomura O, Frank LA (2013) Obelin mutants as
reporters in bioluminescent dual-analyte binding assay. Anal Methods 5:636–640
47. Tannous BA, Verhaegen M, Christopoulos TK, Kourakli A (2003) Combined flash- and
glow-type chemiluminescent reactions for high-throughput genotyping of biallelic
polymorphisms. Anal Biochem 320:266–272
48. Konstantou J, Ioannou PC, Christopoulos TK (2007) Genotyping of single nucleotide
polymorphisms by primer extension reaction and dual-analyte bio/chemiluminometric assay.
Anal Bioanal Chem 388:1747–1754
49. Daly AK (2003) Pharmacogenetics of the major polymorphic metabolizing enzymes. Fundam
Clin Pharmacol 17:27–41
50. Elenis DS, Ioannou PC, Christopoulos TK (2009) Quadruple-allele chemiluminometric assay
for simultaneous genotyping of two single-nucleotide polymorphisms. Analyst 134:725–730
51. Ozava T, Yoshimura H, Kim SB (2013) Advances in fluorescence and bioluminescence
imaging. Anal Chem 85:590–609
52. Xia Z, Rao J (2009) Biosensing and imaging based on bioluminescence resonance energy
transfer. Curr Opin Biotechnol 20:37–44
53. Roda A, Guardigli M, Michelini E, Mirasoli M (2009) Nanobioanalytical luminescence:
Förster-type energy transfer methods. Anal Bioanal Chem 393:109–123
54. Arai R, Nakagawa H, Tsumoto K, Mahoney W, Kumagai I, Ueda H, Nagamune T (2001)
Demonstration of a homogeneous noncompetitive immunoassay based on bioluminescence
resonance energy transfer. Anal Biochem 289:77–81
55. Li F, Yu J, Zhang Z, Cui Z, Wang D, Wei H, Zhang XE (2013) Use of hGluc/tdTomato pair
for sensitive BRET sensing of protease with high solution media tolerance. Talanta
109:141–146
56. Cissel KA, Campbell S, Deo SK (2008) Rapid, single-step nucleic acid detection. Anal
Bioanal Chem 391:2577–2581
57. Kumar M, Zhang D, Broyles D, Deo SK (2011) A rapid, sensitive and selective
bioluminescence resonance energy transfer (BRET)-based nucleic acid sensing system.
Biosens Bioelectron 30:133–139
58. Yamakawa Y, Ueda H, Kitayama A, Nakamune T (2002) Rapid homogeneous immunoassay
of peptides based on bioluminescence resonance energy transfer from firefly luciferase.
J Biosci Bioenq 93(6):537–542
59. Shekhman SS, Ghosh I (2011) Split-protein systems: beyond binary protein-protein
interaction. Curr Opin Chem Biol 15(6):789–797
60. Mie M, Thuy NPB, Kobatake E (2012) Development of a homogeneous immunoassay
system using protein A fusion fragmented Renilla luciferase. Analyst 137:1085–1089
61. Ohmuro-Matsuyama Y, Chung C-I, Ueda H (2013) Demonstration of protein-fragment
complementation assay using purified firefly luciferase fragment. BMC Biotechnol 13:31–39
62. Kim SB, Takenaka Y, Torimura M (2011) A bioluminescent probe for salivary cortisol.
Bioconjug Chem. 22(9):1835–1841
63. Scott SD, Hamorsky KT, Ensor CM (2011) Cyclic AMP receptor protein-aequorin molecular
switch for cyclic AMP. Bioconjug Chem 22(3):475–481
Index
Note: Page numbers followed by ‘‘f’’ and ‘‘t’’ indicate figures and tables respectively
A B
Abortive infection, 159 Bacterial bioluminescent assays, detection of
Acetate kinase, 180 organic compounds using, 114–115
Acinetobacter baylyi ADP1, 123, 124 benzene, toluene, ethylbenzene, and xylene
Acinetobacter bioreporter (BTEX), 115–120. See also BTEX
DF4/PUTK2, 128 bioreporters
DF4-8, 128 alkane aliphatic hydrocarbons, 122–124
Acyl-homoserine lactone (acyl-HSL) synthase, biphenyls, 125–127
46 chlorinated aliphatic hydrocarbons,
AhR nuclear translocator (ARNT) gene/pro- 124–125
tein, 130, 134, 135 phenol and derivatives, 127–128
L M
Laser-induced fluorescence, 184 Mann-Whitney U test, 83
Light emission reaction MCF-7 cell line
catalyzed by bacterial luciferase, 158 b-Globin promoter, 136
catalyzed by firefly luciferase, 158 -derived MELN reporter cell line, 136
Light production. See Bioluminescence -derived MVLN reporter cell line, 136
Light-emitting proteins, 176 MDA-kb2 cell line, 137
Light-producing reaction, 177 bioreporter assay, 140
Listeria, 156, 161, 164 MELN assay, 136, 138, 140, 141
Listeria reporter phage A511::luxAB, 160, 161 reporter cell line, 136, 138
Listeriosis, 164 Mercaptoethanol, 78, 80f
Littoral zone, 19 Microecosystem (MES) components, 91–95
luminous earthworms in, 20 experiments, 93
Locked nucleic acid (LNA), 188, 189 biomass of algae, 94f
Luciferase dynamics of bioassay parameters for,
biotesting, 72–73 94f
scheme, 72f Midwater hatchetfish (Sternoptyx pseudobscu-
biotin-streptavidin/avidin system, 180f ra), 13f
as immunoassay labels, 179–183 Misdirection (or distraction/obstruction of
bacterial luciferases, 179 sight), 4
biotinylated luciferase, 180 Mouse mammary tumor virus (MMTV) pro-
firefly luciferases, 179 moter, 137
immunoprecipitation system (see Lucifer- Multianalytical bioluminescent assay,
ase immunoprecipitation system 189–192
(LIPS)) Mushrooms, 8, 16
luciferase index (LI), 73 attraction of spore-dispersing organisms,
single-chain polypeptides, 181 11
homologous glycosylated, 182 Mycobacterium reporter phages, 162, 165, 166
Luciferase immunoprecipitation system Mycobacterium tuberculosis (MTB) infection,
(LIPS), 181, 181f 185
Luciferin, 4, 15, 21, 28, 118, 138, 179, 189
cypridinid luciferin, 21–22, 22f
substrate luciferin, 158 N
Luminescent secretion clouds, 7 N3-(oxohexanoyl-) L-homoserine lactone
Luminous species (OHHL), 163
brittle star (Ophiopsila), 9f NADH:FMN-oxidoreductase-luciferase, 70,
coastal fish (Acropoma japonicum), 24f 71–72, 76
earthworm (Microscolex phosphoreus), 18f advantages of, 75
freshwater snail (Latia neritoides), 19f in Lake Shira water analysis, 74
marine snail (Angiola zonata), 20f in pond water MES, 91–95
millipede (Paraspirobolus lucifugus), 8, 8f in single system of enzymatic assays, 87
mushroom (Mycena chlorophos), 8, 9f Natural water biotesting
lux genes, 39, 41, 43–47 reference water, Lake Baikal, 84
gene content and gene order of, 44f samples
luxCDABEG, 39, 43 characteristics, 83, 84t
luxF, 43 light emission intensity dependency on,
lux-rib operons, 44, 45 85f
genus-specific differences, 45 toxicity bioassay of, 84t
nonfluorescent flavoprotein coding, 43 toxicological characteristics of, 84t
lux operon expression, regulation of, 47–51 using immobilized coupled enzyme sys-
population density-responsive regulation, tem, 83–85
47 using soluble enzyme system, 83–85
luxRVh (lux operon in V. harveyi), 47 Nucleic acid hybridization assay, 185f
204 Index
S W
(S)-3-hydroxytridecan-4-one (cholerae autoin- Waste water biotesting
ducer, CAI-1), 49 reference water, Lake Baikal, 84
Sakyo Kanda, 27 samples
Salmonella, 162, 167 characteristics, 83, 84t
Salmonella reporter phage Felix O1, 161 light emission intensity dependency on,
Sandwich-type immunoassay, 178f 85f
Shewanella, 39, 40t, 41, 42, 46 toxicity bioassay of, 84t
Shewanellaceae, 39 toxicological characteristics of, 84t
Shigella, 167 using immobilized coupled enzyme sys-
Single-tube luminometers, 161 tem, 83–85
Split-protein reassembly technology, 192 using soluble enzyme system, 83–85
Spore-feeding fungus gnat (Keroplatus nippo-
nicus), 11, 11f
Startling (or deterring), 4 X
click beetles, 6 Xenobiotics, 71
phengodid, 6 contamination of MES, 98
Symbiotic luminescence, 23–24 Xenopus Vitellogenin A2 gene, 136
206 Index
Y Z
Yata Haneda, 27 Zebrafish assay, 134, 137
Yeast-based assays, 138, 139
hAR/ARE-luc bioassay, 139
Yellow fluorescent protein (YFP), 46, 192
energy transfer (EYFP), 192
Yenissei River water analysis, 74
Yersinia pestis, 166