Effects of PH Alterations On Stress - and Aging-Ind
Effects of PH Alterations On Stress - and Aging-Ind
Effects of PH Alterations On Stress - and Aging-Ind
REVIEW
Received: 21 February 2022 / Revised: 26 April 2022 / Accepted: 21 May 2022 / Published online: 24 June 2022
© The Author(s) 2022
Abstract
Upon stress challenges, proteins/RNAs undergo liquid–liquid phase separation (LLPS) to fine-tune cell physiology and
metabolism to help cells adapt to adverse environments. The formation of LLPS has been recently linked with intracellular
pH, and maintaining proper intracellular pH homeostasis is known to be essential for the survival of organisms. However,
organisms are constantly exposed to diverse stresses, which are accompanied by alterations in the intracellular pH. Aging
processes and human diseases are also intimately linked with intracellular pH alterations. In this review, we summarize
stress-, aging-, and cancer-associated pH changes together with the mechanisms by which cells regulate cytosolic pH homeo-
stasis. How critical cell components undergo LLPS in response to pH alterations is also discussed, along with the functional
roles of intracellular pH fluctuation in the regulation of LLPS. Further studies investigating the interplay of pH with other
stressors in LLPS regulation and identifying protein responses to different pH levels will provide an in-depth understanding
of the mechanisms underlying pH-driven LLPS in cell adaptation. Moreover, deciphering aging and disease-associated pH
changes that influence LLPS condensate formation could lead to a deeper understanding of the functional roles of biomo-
lecular condensates in aging and aging-related diseases.
13
Vol.:(0123456789)
and tryptophan [4, 5]. The enrichment of only a few amino condensate formation, eventually providing new opportuni-
acids in these domains results in low complexity that could ties for the prevention and treatment of human diseases.
mediate weak interactions. Other proteins that contain oli- Phase separation of proteins is a multifactor dynamic
gomerization domains and multiple-folded modular domains process, and it occurs not only spontaneously under nor-
are also contribute to the multivalent interactions [5]. Fur- mal conditions, but also upon stimulation from an array of
thermore, emerging roles of RNA molecules in the assem- environmental factors, including changes in temperature,
bly of biomolecular condensates have been revealed. RNAs pH, ATP/energy, macromolecule concentration, and ionic
help establish the promiscuous interaction network through strength [14]. These physiological parameters constitute a
interactions with the RNA-binding domains of proteins, and continuous phase boundary, and crossing this boundary by
their intermolecular interactions and self-assembly define changing one or more parameters, such as by raising the
the compositions of higher-order condensates [6, 7]. Accu- temperature, depriving nutrients, lowering the pH, or chang-
mulating evidence shows that protein post-translational ing other factors, can cause phase separation and the forma-
modification (PTM) is another important mechanism that tion of condensates, which is an adaptive tuned response of
achieves cellular control of protein phase separation and cells [15]. Homeostasis of pH is a prerequisite for the normal
condensate formation through processes such as phospho- survival of organisms. Many proteins are very sensitive to
rylation, acetylation, SUMOylation, ubiquitination, meth- pH alterations, and a very small change in pH can induce
ylation, and ADP-ribosylation [8, 9]. These modifications phase transition of proteins. Phase separation in most pro-
can alter the weak multivalent interactions by changing the teins is triggered at low pH; while in others, it is induced by
charge, structure, hydrophobicity, and other properties of alkaline pH [16]. In vivo, the mechanism by which pH regu-
proteins, thus affecting phase separation behavior [8, 9]. In lates protein phase separation is not completely clear. Here,
addition, not only protein PTM but also RNA PTM affects we review the literature on stress-associated pH fluctuation
condensate dynamics. For instance, N6-methyladenosine in cells, how cells maintain and regulate cytosolic pH, and
(m6A) of RNA can modulate condensate formation and the effects of pH changes on protein phase separation. The
composition by regulating mRNA distribution into distinct mechanisms by which pH can mediate phase separation are
condensates or changing the phase separation behaviors of also discussed. Further research on these topics will not
its binding partners [10, 11]. only advance our understanding of compartment forma-
There is mounting evidence that LLPS and condensate tion affected by pH changes but will also provide important
formation are widely present in the cells and play important insight into the relationship between pH and a diverse set of
roles in a wide range of physiological processes (Fig. 1), human diseases.
including chromatin organization, cytoskeletal assembly,
signal transduction, transcriptional regulation, protein deg-
radation, cell division and differentiation, and environmental Diverse stresses induce intracellular pH
response and adaptation [2]. The condensates can transition fluctuation
into different material states such as gel- or solid-like states
[1, 12]. Therefore, maintenance of normal condensate mate- Cytosolic pH is a tightly controlled physiological param-
rial properties can ensure the assembly and disassembly of eter in all cellular systems, as almost all cellular processes
condensates in a tightly controlled manner to fulfill origi- depend on a constant pH for normal functions [17–21].
nal functions, while aberrant phase transition is causatively For instance, in yeast, pH is involved in replicative senes-
associated with the onset and development of age-related cence of mother cells and rejuvenation of nascent daugh-
neurodegenerative diseases and cancers (Fig. 1) [13]. In ter cells [22], and cytoplasmic acidification is critical for
recent research, the development of methods to study LLPS yeast cells to enter dormancy under stress conditions [23].
has become an important objective. A series of tools have In plants, intracellular pH changes are components of a
been developed to predict and analyze the phase separa- number of phytohormone signaling pathways, modulat-
tion capabilities of proteins [1]. Fluorescence microscopy ing gene expression and defence [21, 24]. In mammals,
observation techniques, including fluorescence recovery the maintenance of pH homeostasis is of key importance
after photobleaching (FRAP) and superresolution imaging, for the proper execution and regulation of neurotrans-
can also provide more detailed information on the material mission [25]. Small changes in cytosolic pH can lead to
properties, composition, and dynamics of biomolecular con- major changes in metabolism, signal transduction, protein
densates. In addition, in vitro reconstitution using purified folding, and protein–lipid interactions [19, 20]. However,
proteins is an accessory method used for studying LLPS [1]. organisms are often exposed to diverse adverse conditions
These methods can help researchers to further elucidate the throughout their life cycles, and stress-induced cytosolic
compositions of biological molecules and related biologi- pH fluctuations are broadly present in the cells; these fluc-
cal reactions and explore the factors that drive or influence tuations are induced, for example, by osmotic stress, heat
13
Fig. 1 Liquid–liquid phase separation in mammalian cells and its LLPS. a Mutations in the substrate recognition domain of the tumor
involvement in aging-related neurodegenerative diseases and can- suppressor SPOP prevent its binding to oncogenic substrates and sub-
cers. Under physiological conditions, scaffold biomacromolecules sequent condensate formation with ubiquitin ligase complex, causing
undergoing liquid–liquid phase separation (LLPS) can interact with a failure of oncogenic substrate ubiquitination and proteasomal deg-
and recruit other client molecules to form reversible liquid-like con- radation. b Mutation of p53 can accelerate its solid-phase transition
densates, which participate in a wide range of physiological pro- into amyloid aggregates, which is found in more than 50% of human
cesses. During aging, multiple factors, including protein mutation and cancers. c Chromosomal translocations lead to aberrant condensate
repeated expansions, cellular environmental and metabolic changes, formation of transcriptional regulators (TRs) at enhancers and pro-
damage to protein quality-control systems, and abnormal protein moters of oncogenes, driving abnormal oncogenic transcriptional pro-
localization and post-translational modification, can affect the LLPS grams. d Mutation or overexpression of signaling receptors alter the
process and promote aberrant gel-like condensate or pathological pro- formation of signaling clusters and activates aberrant signaling cas-
tein aggregate formation, ultimately leading to the onset and progres- cades, contributing to cancer development
sion of neurodegenerative diseases. Tumorigenesis is also related to
13
13
Stress Species pHi Effects of stress The reason of pH change References
Page 4 of 23
Heat shock Yeast ↓ Membrane permeability increase Protons in the environment influx [19, 30, 33–35]
Drosophila Slight cell swelling and altered metabolic activ- Intracellular protons increase [28, 36, 37]
ity
Mammals Inhibit cell growth and change plasma mem- Inhibition of N
a+–H+ exchange and metabolic [38–42]
brane fluidity, permeability to small molecules, pathways
and membrane-bound enzyme activity
Starvation Yeast ↓ Decrease cytoplasmic mobility and volume, and Energy shortage to pump protons out [23, 43–47]
cell enters dormancy
Plasmodium Block mitosis Lack of energy to maintain the pH gradients [48]
inside and outside the cell
Osmotic stress Bacteria ↓ Change cell volume and metabolic processes Cell loses water and the concentration of protons [49, 50]
increases, and activate distinct OmpR-related
pathways
↑ Proton efflux and osmolarity-stimulated K
+ [51–53]
uptake
Protists ↓ Cells shrink, largely rearrange cellular proteins Cell loses water and secretes protons [54–56]
between compartments and decrease activity
Weak acid Yeast Bacteria ↓ Decrease cell growth rate and cell growth stasis Intracellular protons increase and inhibit the [45, 46, 57–59]
ability of cells to maintain normal pH
Hypoxia and anoxia Mammals Plants ↓ Cytoplasmic acidosis or cell death Metabolites produced by anaerobic fermenta- [26, 29, 60–62]
tion/ respiration, such as lactic acid
Alcohols Yeast ↓ Interfere with membrane transport by changing Proton permeability increase [63, 64]
the lipid composition of the plasma membrane
Pathogen Plants ↓ Cause pathological damage and activate defense Oxidation burst [21, 65]
responses
Light intensity Plants ↑ (light enhance), ↓ (light reduce) Affect photosynthesis Proton entering/leaving the thylakoids [21, 66]
Aging Yeast ↑(vacuole) Cells lose their physical integrity, resulting in Pma1 accumulates on the plasma membrane [22, 67]
Mammals (Rat ↓ impaired function (such as mitochondria/lyso- Na+–H+ exchange may be impaired [32]
hippocampus) some dysfunction) and increased risk of death
or diseases
Oxidative stress Plasmodium ↓, Lose pH control and decrease intracellular ATP Inhibition of V-ATPase activity [68]
↑ (vacuole) level
folded proteins and aggregates [70]. On the one hand, upreg- to changes in membrane permeability [90]. In turn, heat-
ulated chaperones can efficiently refold nonnative proteins induced proton influx and pH decreases can activate plasma
or promote the degradation of protein aggregates through membrane ATPase, whose activity is necessary for cell
autophagy or the ubiquitin–proteasome system (UPS). On survival under heat shock [33, 91]. The plasma membrane
the other hand, they can regulate the deposition of certain ATPase pumps intracellular protons out of the cell, partially
misfolded proteins into specialized cellular locations to offsetting the internal acidification resulting from the heat-
shield them from degradation and to refold them after stress induced increase in membrane permeability [33]. In mam-
[71]. In addition to HSR, another adaptive mechanism called malian cells, heat shock leads to a dramatic loss of plasma
the unfolded protein response induced by endoplasmic retic- membrane Na+–K+ ATPase activity, which then results in
ulum stress is also activated upon heat exposure and helps to loss of the inwardly directed electrochemical Na+ gradient
mitigate the damage caused by heat [72, 73]. Moreover, in across the membrane [39]. Therefore, it is speculated that
different research models, ubiquitination-dependent [74–77] Na+ gradient-dependent H+ export from the cytoplasm to
and autophagy-dependent degradation [78–82] have been the outside by Na+–K+ ATPase is affected and that the cyto-
observed to be induced after heat shock, and the activation plasm is acidified during heat stress.
of these degradation systems is essential for cell survival
and recovery from thermally induced protein aggregation Starvation
[74, 82].
In addition to the activation of evolutionarily conserved A decrease in cytosolic pH can also be caused by starva-
systems that contribute to thermotolerance, temperature tion. In yeast cells, numerous studies have shown that
change is often coupled with fluctuations in cytoplasmic pH the cytoplasmic pH decreases from approximately 7.4 to
[28, 83]. Some studies have shown that heat shock acidifies approximately 6.0 under starvation conditions [43, 45, 47].
the cytoplasm. For instance, in yeast, an intracellular pH Pma1, the plasma membrane-localized P-type H+-ATPase
drop can be induced by heat shock [35]. The same heat-asso- in yeast, is involved in pumping protons out of the cells and
ciated pH changes have also been observed in Drosophila is a primary contributor to the maintenance of cytosolic pH
melanogaster [28] and rat hepatoma cells [41]. Stress-asso- stability near neutrality [92, 93]. Importantly, its activation
ciated acidification is thought to be toxic to cells in some requires glucose-regulated phosphorylation [94]. In addition,
cases [29, 84]; whereas in other cases, it might be a cyto- other pumps, such as V-type H+-ATPases (V-ATPases), are
protective strategy that promotes cellular fitness under stress also responsible for cytosolic pH regulation [95]. They work
[23, 33, 85]. For example, cytosolic acidification is required by pumping excess protons into the vacuole to regulate cyto-
for HSR induction in translationally inhibited cells under solic pH homeostasis; they also maintain effective localiza-
heat shock, which allows the cells to adapt to high tempera- tion of Pma1 at the plasma membrane [95, 96]. Glucose is
ture by increasing the transcription of quality-control com- also required for V-ATPase activation because it mediates
ponents [35, 86]. Furthermore, some stress granule (SG) reversible associations between the V1 and V0 domains of
resident proteins, such as DEAD-box RNA helicase Ded1 V-ATPase [43, 97]. Under favorable conditions (with glu-
[87], poly(A)-binding protein 1 (Pab1) [34], and poly(U)- cose), V-ATPase cooperates with Pma1 to pump protons
binding protein 1 (Pub1) [88] in yeast, and Ras-GTPase- out of the cytoplasm and help cells stabilize cytoplasmic
activating protein SH3-domain-binding protein (G3BP1) pH in an ATP-dependent manner. However, upon glucose
in mammalian cells [89], have been reported to respond to depletion, a drop in cytoplasmic pH is observed, as starved
elevated temperatures to undergo LLPS. Likewise, they can yeast cells lack efficient H+-ATPase assembly and activation
also respond to low pH that mimics the pH conditions during to support the proton gradient across the membrane [46].
heat stress. Therefore, heat-induced acidification may play a Intracellular protons cannot be discharged outside of the
key role in protein LLPS following heat exposure and then cell. Instead, they accumulate inside the cell; thus, cytosolic
regulate SG dynamics and cell survival under or after stress. pH decreases. The increased concentrations of intracellular
The mechanism by which heat shock acidifies the cyto- protons cause the phase transition of the cytoplasm from a
plasm is not fully understood. However, evidence has shown fluid-like to a solid-like state, and such a dormant or qui-
that the compositions and structures of the cell membrane escent state is a protective strategy for cell survival under
are very sensitive to changes in temperature. In yeast, heat conditions of starvation [23]. Likewise, evidence suggests
shock increases membrane permeability, resulting in pro- that nutrient supply is also closely related to cytoplasmic
ton influx and a rapid decrease in intracellular pH [90]. pH in Physarum plasmodium. The cycle of intracellular
Studies have shown that intracellular pH disturbance is the pH corresponds to the period of the cell cycle of P. plas-
triggering mechanism of thermotolerance in yeast [33], modium. When P. plasmodium is growing in non-nutrient
and changes in plasma membrane compositions contribute medium, the intracellular pH remains stable and then begins
to the thermotolerance of cells, which may also be related to decline gradually, which serves to block normal mitosis.
13
However, upon refeeding of starved P. plasmodium with the human body is often disturbed [115]. Thus, normal osmotic
nutrient medium, intracellular pH can recover to normal val- regulation is impaired, and this impairment is followed by
ues and the cell cycle resumes [48]. increases in the incidence and severity of diseases, such as
hypoosmolality and hyperosmolality [116].
Osmotic stress Interestingly, a growing body of evidence implicates
hyperosmotic stress as a factor leading to internal pH alter-
In addition to heat shock and starvation, osmotic stress is ation. In Listeria monocytogenes, a ubiquitous gram-pos-
another important environmental factor affecting cell sur- itive food-borne pathogen, the initial response to osmotic
vival and growth. Organisms including microbes, plants, stress caused by sorbitol or NaCl is a decrease in intracel-
and mammals, are commonly confronted with hyperos- lular pH [50]. Hyperosmotic stress also leads to cytosolic
motic conditions, which trigger a series of actions resulting acidification in Dictyostelium discoideum, which works as
in downregulation of cellular activity and progression of a novel signal mediator responsible for hyperosmotic stress
disease [98–100]. When osmolarity changes, cells adjust responses [54]. Moreover, another study has indicated that
their volumes accordingly in response to the changing envi- hyperosmotic shock elicits a transient increase in Escheri-
ronment. Cells mainly regulate volume changes by control- chia coli cytoplasmic pH, but the pH returns to normal val-
ling substance influx and efflux, which is usually manifests ues after osmotic adaptation [52]. However, whether and
as cell contraction or expansion, so that cells can return to how osmotic dysregulation in mammalian cells alters pH
a normal resting state [101–103]. A variety of membrane and whether it is relevant to human diseases remain unclear;
transporters are involved in this complex regulation process. thus, these aspects require further investigation to advance
For example, in a hypotonic environment, mammalian cells our understanding of pH-related condensate formation and
initially expand via water uptake and subsequently undergo diseases.
compensatory shrinkage to partially regulate volume reduc- Taken together, the evidence indicates that diverse envi-
tion, usually through efflux of KCl and organic osmolytes ronmental alterations contribute to intracellular pH fluctua-
[104, 105]. In contrast, in hypertonic environments, cells tion. Manipulating intracellular pH not only serves to main-
undergo transient dehydrating contraction by absorbing tain the morphology and function of cells to ensure normal
Na+, K+ and CI− and then pumping out N a+ to regulate the growth and metabolic activities, but also is associated with
increase in cell volume [104]. the preservation of cellular equilibrium in response to sev-
Intracellular osmotic homeostasis is necessary to maintain eral environmental factors, which could promote cellular
normal cell function and survival, and osmotic dysregulation fitness.
is the basis of many diseases and their complications, includ-
ing cataracts [106], epilepsy [107], inflammation [100, 108], Aging and neurodegenerative diseases
and hypernatremia [109]. For instance, in hyperglycemia
or hypergalactosemia, activated aldose reductase converts Aging is usually an irreversible biological process and is
glucose and lactose to galactose and sorbitol, respectively, considered to be a predominant risk factor for many neuro-
which accumulate in the lens and cause osmotic swelling, degenerative diseases [117]. Nine hallmarks of aging have
leading to diabetic cataracts [106]. In addition, cancer and been tentatively identified in different organisms, including
aging processes are also closely related to intracellular genomic instability, telomere attrition, epigenetic altera-
osmotic regulation. Many studies have shown that ion chan- tions, loss of proteostasis, deregulated nutrient sensing,
nels and ion pumps are beneficial to the development and mitochondrial dysfunction, cellular senescence, stem cell
progression of cancer [110]. Given the importance of ion exhaustion, and altered intercellular communication. These
channels for osmotic homeostasis and the abnormal expres- hallmarks can be classified into three layers: primary hall-
sion of transporters in many cancers [111], it is likely that marks, antagonistic hallmarks, and integrative hallmarks,
the original homeostasis in cells will be disrupted, creating which co-occur during aging and are usually interconnected
a more favorable internal environment for cancer develop- with each other; defining the exact relationships and causal
ment. Interestingly, Yes-associated protein (YAP), is a tran- network of these hallmarks may contribute to future studies
scriptional coactivator that is widely activated in cancer cells on aging and aging-related diseases [118].
[112], can sense the tumor microenvironment and modify In addition to the hallmarks of aging discussed above,
the physicochemical properties of the surrounding environ- growing evidence shows that intracellular pH alterations are
ment by activating transcription, thereby promoting tumor also intimately linked to aging processes and aging-related
development [113]. Moreover, YAP-activated transcription neurodegenerative diseases. In mammals, the intracellular
is mediated by the LLPS process, which also occurs under pH of central neurons is tightly regulated, and its fluctua-
hypertonic conditions [114]. Aging does not directly cause tions are important for signaling and synaptic plasticity [119,
disease, but in this process, the homeostasis of water in the 120]. Specifically, in cortical neurons, a mild intracellular
13
pH decrease occurs following an excitability increase, and (Fig. 2), which utilizes the inwardly directed electrochemical
this decrease acts as feedback to reduce local bioelectric Na+ gradient generated by Na+–K+ ATPase to export H+
activity and excitability. However, when the intracellular pH [32]. Moreover, limited ATP synthesis during aging might
is outside a certain range and reaches its limits, there may be also affect ATP-driven ion pumping, including N a+ gradi-
+ +
an increased risk of cell death [25, 119, 121, 122]. Impor- ent generation by N a –K ATPase [131]. The impacts of
tantly, a decrease in neural pH levels has been observed in a aging-related alterations on pH regulation are controversial.
number of neurodegenerative disorders [123, 124] and even A slight decrease in intracellular pH may provide neuropro-
in the normal aging process [32, 125, 126]. Moreover, acute tection [132], while successively greater acidification may
neuroinflammation has been observed to provoke intracellu- increase the vulnerability of brain tissue to stressful condi-
lar acidification in the mouse hippocampus [127]. For exam- tions [125, 133–135].
ple, in mammalian cortical neurons, intracellular pH is nega- In addition to cytosolic pH dysregulation, lysosomal/
tively correlated with aging, as evidenced by significantly vacuolar pH dysregulation has also been implicated in aging
lower pH in hippocampal slices from aged rats than in slices and aging-related neurodegenerative diseases. Evidence is
from young rats [32, 128]. Likewise, in human neurons, the now emerging that defective lysosomal function is a major
intracellular pH has also been observed to decrease with factor in the pathogeneses of different types of neurodegen-
aging [126, 129]. The mechanisms involved in decreased erative diseases, specifically, a failure of the maintenance
intracellular pH may be the disruption and overwhelmed of a highly acidic lysosomal/vacuolar pH [136]. There is
of pH regulatory systems through processes including also increasing evidence for aging-related compromise of
aging-related decreases in buffering capacity and disrup- lysosomal function [22]. In yeast, vacuolar pH is a critical
tion of diverse transmembrane acid/base-transporters [32, regulator of mitochondrial function and replicative lifespan.
128–130]. For instance, considering that Na+–H+ exchange Vacuolar acidity declines with aging, and reduced vacuolar
is the dominant regulatory mechanism for proton extru- acidity disrupts pH-dependent amino acid homeostasis in
sion in cultured hippocampal neurons, altered H+ homeo- the vacuolar lumen, resulting in age-related dysfunction
stasis might be attributable to impaired Na+–H+ exchange of mitochondria and a shortened lifespan [67]. In addition,
Fig. 2 Aging affects intracellular pH. When cells are young, P-type pH decreases. Moreover, cell buffering capacity is also impaired dur-
H+-ATPases distributed on the plasma membrane act in concert ing aging. V-type H+-ATPase is a target of oxidative stress in aging.
with V-type H+-ATPases localized on the lysosomal/vacuolar mem- Increased oxidative modification of V-type H+-ATPase might inhibit
brane to regulate intracellular pH. However, during aging, for exam- V-type H+-ATPase-mediated vacuolar acidification. Alternatively,
ple, in yeast, P-type H+-ATPase Pma1 accumulates on the plasma aging might alter lysosomal/vacuolar acidification by downregulat-
membrane, and excessive H + is pumped out of the cell, resulting in ing V-type H+-ATPase subunit expression, lowering the availability
reduced cytosolic H availability for V-type H+-ATPase. This leads
+
of V-type H +-ATPase. The solid lines represent normal ion transport.
to a decrease in vacuolar acidity. In other cases, such as in the aged The dashed lines represent impaired ion transport. In the young cell
rat hippocampus, the Na+–K+ pump and N a+–H+ exchange may be cytoplasm, yellow represents cytoplasm with a normal pH. In the
impaired; as a result, H+ accumulates in the cytoplasm, and cytosolic aged cell cytoplasm, red represents cytoplasm with a decreased pH
13
13
hand, the increased intracellular pH can increase cell prolif- of distinct transport pathways. For example, P-type proton
eration, facilitate apoptosis evasion, and promote cytoskel- pumps are widely distributed on eukaryotic cell membranes,
etal remodeling for cell migration. On the other hand, the and they are the main determinants of proton efflux and cyto-
acidified extracellular environment can increase the activi- plasmic pH control in plants and yeast [19, 173]. As mentioned
ties of acid-activated proteases and promote extracellular above, Pma1 is the most abundant protein in the plasma mem-
matrix degradation, thereby accelerating tumor cell invasion brane of yeast and actively coordinates with V-ATPases to
and dissemination [31]. However, during these processes, regulate cytosolic pH [19, 173]. V-ATPases can also acidify
whether and how pH alterations of cancer cells are related compartments in an ATP-dependent manner and are distrib-
to the aberrant phase behavior of cancer-related proteins or uted in acidic organelles such as the Golgi apparatus, vacuole/
aberrant formation of membrane-less compartments such lysosomes, and endosomes of all eukaryotic cells [180]. In
as SGs, PML bodies, paraspeckles, and amyloid bodies, yeast cells, V-ATPase activity is indispensable for vacuolar
remains unclear and requires further in-depth investiga- acidification during glucose metabolism and homeostasis of
tion. Such research will provide more knowledge about the cytoplasmic pH in the short term. In the long term, V-ATPase
molecular basis of cancer and facilitate the development of is very important for the stability of Pma1 localization [95].
new therapies. F-type proton pumps are mainly distributed in the bacterial
plasma membrane, the mitochondrial membrane, and the plant
endomembrane. In enterococci, when the cytoplasm is acidi-
Cytosolic pH control by metabolism‑based fied, the level and activity of F-type H+-ATPase increase syn-
and transporter‑based regulation chronously, leading to cytoplasmic alkalization [181]. When
the pH value is restored to the initial value, the decrease in the
Since pH control is a critical requirement for growth in all amount and activity of the enzymes terminates proton extru-
organisms, different organisms have adopted a number of sion. Thus, changes in the amount and activity of enzymes
common strategies to address the challenges of pH main- seem to be necessary for pH regulation [182].
tenance in the face of rapid metabolism and extracellular Moreover, these proton pumps act in concert with a large
environment changes [171–173]. Cells separate metabolites, array of other transporters. Increasing evidence indicates
proteins, and biochemical processes in a manner dependent that a number of ion/H+ exchangers are also important for
on compartmentalized membrane-bound organelles, each intracellular pH regulation in different organisms, including
of which has distinct pH requirements and pH regulation yeast, plants, and mammals [19, 172, 173]. These exchang-
mechanisms [172]. More importantly, cellular compartments ers couple the transfer of H + across biological membranes to
have inherent pH buffering capacities. This buffering is counter-transport of other cations, such as N a+ or K
+, to pro-
achieved by the presence of various intracellular weak acids tect against excess acidification. Furthermore, Na+-coupled
and bases, as well as the ionizable groups of macromol- HCO3− transporters, which are involved in the uptake of extra-
ecules such as side chains of amino acids [174]. Moreover, cellular HCO3−, have also been reported to play key roles in
cytosolic pH regulation also relies on metabolites produced the regulation of cytosolic pH. They contribute to the main-
by pH-dependent biological reactions [175]. Organic acids tenance of C O2–HCO3− equilibrium, the most important pH
such as malate can produce or consume H + via carboxylation buffering system [183, 184]. Although the importance of pro-
and decarboxylation reactions. Therefore, correct synthe- ton extrusion in pH control has been revealed, acid-importing
sis, degradation, and transport of organic acids through the transporters such as C l−–HCO3− exchangers, which allow
−
cytoplasm to other organelles are thought to be important HCO3 efflux, can efficiently prevent overalkalization of the
strategies to regulate intracellular pH [176, 177]. In addition, cells by working counter to C O2–HCO3− transporters to enable
the alternative pathways to glycolysis, the cyanide-resistant the fine control of cytosolic pH [185].
alternative respiration pathway and malate-derived lactic and In summary, cells exhibit a complicated pH regulation net-
alcoholic fermentation, which are unique to plants, jointly work dependent on the interplay among multiple transporters
regulate pH homeostasis in plants [175]. In mammalian cells that import or export proton equivalents and metabolism-based
and fermenting yeast, CO2 produced during metabolism can regulatory mechanisms, and this network can accurately regu-
diffuse freely through biological membranes. It can react late and maintain cytosolic pH. More details can be found in
with water to form H CO3−, which is an effective proton recent reviews [19, 171–173].
buffer and consumes protons to produce carbonic acid when
the cells are confronted with an acute drop in intracellular
pH [178, 179].
However, when cells are under long-term stress, the major
regulatory mechanism to maintain cytosolic pH homeostasis
is the membrane transport of H+, which involves a large array
13
pH‑dependent phase separation condensate switch in SG assembly. Its phase separation also occurs in
formation induced by stress an RNA-dependent manner under low pH and heat shock
[89, 203]. Moreover, members of the Asp–Glu–Ala–Asp
As discussed above, many types of stress cause a decrease (DEAD)-box ATPase (DDX)3 family are widely present
in cytoplasmic pH, and these stress conditions are known in both eukaryotes and prokaryotes [192, 194], and stud-
to induce phase separation of proteins to form conden- ies have suggested that many proteins in the DDX family
sates. Here, we summarize the proteins that are known to undergo LLPS in vivo or in vitro, including Ded1, Dbp1,
form phase separation condensates in response to pH stress and Dbp2 in yeast; DDX3X, DDX4, and DDX6 in humans;
together with other stresses in which phase separations are and DeaD, SrmB, and RhlE in E. coli [192, 193]. Ded1p,
mainly affected by pH alterations, such as heat shock and an ATP-dependent DEAD-box RNA helicase in yeast, is
starvation (Table 2). an indispensable translation initiation factor and a compo-
Many biomolecules undergo LLPS to form liquid-like nent of SGs [188]. It can parse the secondary structure of
condensates that mediate diverse cellular functions [222, mRNA 5′ untranslated regions for ribosomal scanning and
223]. For example, autophagosome formation is a pro- recognition of the initiation codon [186, 187]. Studies have
cess that is precisely regulated by protein phase separa- shown that Ded1p undergoes phase separation and forms
tion. Atg1 complex formation is a prerequisite for preau- condensates at elevated temperatures, or at lower tempera-
tophagosomal structure (PAS) assembly and autophagy tures when the pH is adjusted to that of the heat-stressed
initiation [202]. Recent research suggests that the PAS is cytosol (heat-shocked cells experience a decrease in cyto-
a liquid-like condensate formed by phase separation of solic pH). When in condensate form, Ded1p is translation-
the Atg1 complex, which is critical for further dynamic ally inactivated, which leads to a switch in translation from
recruitment of other proteins or factors during autophago- housekeeping transcripts to stress-responsive transcripts
some formation. Notably, this process occurs under low [87]. Therefore, heat shock-induced and temperature-asso-
pH and starvation conditions [200–202]. TORC1 is a ciated pH change-induced Ded1p condensation in SGs is
modulator of PAS organization that targets Atg1 complex an adaptive response to survive heat shock. It promotes an
assembly by regulating the phosphorylation/dephospho- evolutionarily conserved heat shock response that selec-
rylation of Atg13, a component of the Atg1 complex [202, tively translates housekeeping or heat shock transcripts
224]. Its activity is also modulated by phase-separated [87]. Similarly, another DDX family member in yeast,
compartments such as SGs. Under stressful conditions, Dhh1, is responsible for the assembly and disassembly
including heat, starvation, and osmotic stress, TORC1 is of RNA-containing membrane-less organelles. Dhh1 also
recruited into SGs; as a result, TORC1 signaling is inhib- exhibits enhanced phase separation at low pH, which mim-
ited [225–227]. For example, in yeast, TORC1 is parti- ics the pH conditions during glucose starvation [192].
tioned into heat shock-induced SGs, which then prevents Moreover, evidence indicates that in changed growth
an increase in the frequency of heat-induced DNA muta- conditions, enzyme activities can be acutely regulated
tions [225]. Under osmotic stress, TORC1 in mammalian through the formation of phase separation-induced enzyme
cells is similarly sequestered into SGs, thereby blocking its condensates, which restrict or promote specific biochemi-
signal transduction to downstream effectors [227]. cal reactions in membrane-less organelles, suggesting the
SGs are also dynamic membrane-less organelles, the importance of phase separation in regulating the metabo-
formation of which is driven by LLPS [228, 229]. It has lism of cells [47, 195]. For instance, glutamine synthetase
been reported that many proteins in SGs exhibit phase (Gln1) is an indispensable metabolic enzyme that catalyzes
separation behavior under stress-associated pH changes. the synthesis of glutamate and ammonium into glutamine,
Pab1, is an RNA-binding protein consisting of a short a process that requires ATP. Gln1 forms filaments during a
N-terminal sequence, four RNA recognition motifs state of advanced cellular starvation, and filament formation
(RRMs), a proline-rich low-complexity region (LCR) and leads to enzymatic inactivation [197]. Further evidence dem-
a C-terminal peptide-binding domain. It plays a key role onstrates that starvation-induced cytosolic acidification is
in controlling the polyadenylation, stability, and transla- the trigger for Gln1 condensate formation, and many meta-
tion of mRNA in yeast cells [34, 190]. Pub1 is similar bolic enzymes follow this principle to help cells endure and
to Pab1 in that it is an RNA-binding protein with three recover from severe starvation conditions [47].
RRMs and one LCR [191]. Both Pub1 and Pab1 are core In addition to the above-mentioned findings, there are
components of SGs and are prone to phase separation other proteins for which LLPS is directly or indirectly
when temperature increases, pH decreases, or nutrients affected by pH changes, increasing cell fitness or inducing
are lacking to help cells survive during stress [34, 88, diseases. For instance, Sup35 is a translation termination
189]. In addition, G3BP1 is a central node and molecular factor in budding yeast [198]. It can form condensates upon
energy depletion or at a low pH. This pH-dependent phase
13
Ded1p Initiate translation IDR + Yeast Acidic pH, heat shock − [87, 186–188]
Pab1 Control mRNA polyade- LCR, RRMs − Yeast Acidic pH, heat shock Act as signal messenger [34, 189, 190]
nylation, stability, and and affect electrostatic
translation interaction
Pub1 Regulate translation LCR, RRMs − Yeast Acidic pH, heat shock, Affect protein solubil- [88, 191]
glucose starvation ity and electrostatic
interaction
DDXs Coordinate mRNA LCDs + , − (if excess) Bacteria, Yeast, Mam- Acidic pH, glucose − [192–194]
de-capping and decay, mals starvation
regulate general trans-
lational repression
Gln1 Promote the conver- − − Bacteria, Yeast Acidic pH, glucose Act as signal messenger [47, 195–197]
sion of glutamate into starvation
glutamine
Sup35 Terminate translation The N-terminal prion − Yeast Acidic pH, glucose Act as signal messenger [44, 198]
domain and the elec- starvation
trically neutral domain
Atg1 complex Participate in PAS IDRs − Yeast Acidic pH, glucose Possible act as signal [199–202]
assembly starvation messenger
Effects of pH alterations on stress‑ and aging‑induced protein phase separation
G3BP1 Promote SG assembly IDRs, nuclear transport + Mammals Acidic pH, heat shock, Affect protein solubil- [89, 203]
and inhibit RNA factor like domain, osmotic stress ity and electrostatic
aggregation RBD interaction
SARS-CoV-2 N protein Participate in viral RNA IDR1 + Virus Acidic pH Affect electrostatic [204–206]
replication and virion interaction
packaging
α-Syn Act as a SNARE- The N-terminal region − Mammals Acidic pH Affect electrostatic [149, 207–209]
complex chaperone (most family disease interaction
and contribute to mutations occur) and
Parkinson’s disease the “non-amyloid-β
pathogenesis component” region
4R-Tau Induce tubulin assembly The microtubule-bind- − Mammals Lower-critical solution Affect electrostatic [16, 210–212]
and stabilize micro- ing repeats transition interaction
tubules
FUS Participate in DNA LCDs + , − (high ratios) Mammals Acidic pH, DNA dam- − [213, 214]
13
Page 11 of 23 380
380
Page 12 of 23 X. Jin et al.
[217–221]
of the yeast cell from stress [44]. The nucleocapsid protein
(N) of the severe acute respiratory syndrome coronavirus
(SARS-CoV-2) is a multivalent RNA-binding protein that
is essential for viral RNA replication and virion packag-
solubility and electro-
Possible affect protein
ing [206]. The N protein can partition into SGs and inter-
static interaction
13
disruption of condensate formation leads to destabilization in response to these stresses [34, 88]. It is known that LLPS
of p53 and reduced induction of its target genes as well as is driven by multivalent weak macromolecular interactions
cell cycle arrest [151]. Interestingly, it has been reported (protein–protein, protein–RNA, and RNA–RNA interac-
that 53BP1 can respond to low pH to form 53BP1 droplets tions), disruption or alteration of which would affect pro-
[151]; thus, further studies on the relationships of pH regu- tein phase separation behaviors [5, 233]. Therefore, pH
lation and 53BP1 LLPS will help enhance our understand- changes can influence intramolecular or intermolecular pro-
ing of tumorigenesis. However, besides 53BP1, research on tein–protein/RNA interactions by changing the net charges
the relationships between cancer-associated proteins and of components, thereby driving LLPS (Fig. 3). For example,
pH dysregulation are limited. Considering that the physi- G3BP1 is a multidomain protein composed of two folded
ochemical properties and microenvironment of cancer cells domains and two IDRs. Under nonstress conditions, its
are different from those of normal cells [168], two impor- central negatively charged, glutamate-rich IDR can interact
tant research topics in the future are whether these proteins with the C-terminal positively charged RG-rich region to
undergo pH-regulated LLPS and how pH-regulated LLPS allow G3BP1 to fold into a compact state. This compact
is relevant to tumorigenesis. In addition, research on how state is an autoinhibitory conformation that disrupts G3BP1
microenvironmental changes in cancer cells affect the phase separation. However, at a low pH, protonation of the
dynamics of intracellular membrane-less organelles such as clustered glutamates changes the net charge of the acidic
SGs, PML bodies, paraspeckles, and amyloid bodies, whose IDR and disrupts its stable electrostatic interactions with
aberrant assembly is associated with cancer, is also needed. the RG-rich region, allowing G3BP1 to expand from its
Such research will provide further evidence regarding the original tightly self-inhibited state and release the RG-rich
links among pH, LLPS, and cancer. region. G3BP1 can then further facilitate intermolecular
protein–RNA/protein interactions to drive LLPS, which is
consistent with the observation that heterotypic interactions
Mechanisms underlying pH‑mediated phase among G3BP1 and RNA molecules drive SG assembly [89,
separation 203]. In addition, a low pH can directly trigger Pub1 assem-
bly, and this pH-dependent assembly formation is sensitive
pH changes mediate protein–protein/RNA to salt concentrations, suggesting that electrostatic interac-
interactions tions promote Pub1 assembly. Self-interactions among the
RRM domains are the main drivers for Pub1 phase sepa-
Some proteins or their specific domains possess the ability ration, and acidic pH may change the charge distribution
to sense stresses directly and thus undergo phase separation in the RRM domains, thereby mediating the electrostatic
Fig. 3 Roles of pH in biomolecular condensate formation. Under multiple functional roles in triggering liquid–liquid phase separation
nonstress conditions, proteins and RNAs are dispersed in the cyto- (LLPS)-driven condensate formation; for example, it affects protein–
plasm. When cells are exposed to stresses such as starvation, heat protein/RNA interactions, alters protein solubility, or acts as a mes-
shock, or acid stress, the intracellular pH changes, and this change senger to transmit stress signals. pH changes can also enhance phase
is accompanied by the formation of protein- and RNA-containing separation, which may gradually mature and result in transformation
biomolecular liquid-like condensates. During this process, pH plays into an irreversible gel-/solid-like state
13
interactions [88]. Moreover, the pH range that can induce changes in pH can affect the solubility of Pub1 and lead to
artificially recombinant ELP phase separation is related to the formation of stress-responsive Pub1 condensates [88].
the pKa. This suggests to a certain extent that pH can affect Changes in pH appear to be able to decrease the solubili-
protein–solution or protein–protein interactions by changing ties of many proteins. In yeast, a decrease in pH results in a
the number of ionizable residues of proteins, thus trigger- phase transition of cytoplasm from a fluid-like to a solid-like
ing phase separation [217]. Likewise, phase separation of state, which might be caused by decreased solubilities of a
Pab1 at low pH is also an electrostatically mediated process series of proteins and subsequent formation of intracellu-
[34]; thus, the principle of pH-dependent protein condensate lar solid-like assemblies, such as SGs [23, 235]. Moreover,
formation mediated by electrostatic interactions may be gen- the relationship between pH and protein isoelectric point is
eralizable to many proteins. closely related to solubility. The closer the pH value is to the
Notably, pH changes not only initiate protein LLPS by isoelectric point of the protein, the lower its solubility, and
facilitating intramolecular or intermolecular interactions the more likely it is that phase separation occurs [236, 237].
but also enhance phase separation and its further maturation This could explain to a certain degree why the microtubule-
into a gel state or a pathological solid state (Fig. 3). Phase binding repeats of Tau are most prone to phase separation
separation of α-Syn is mediated by an interplay of electro- when the pH is close to the isoelectric point but less prone
static interactions in the unstructured N-terminal domain to demixing when the protein solubility is increased in
and hydrophobic interactions in the central NAC domain, response to pH that is substantially higher or lower than the
while the charge distribution in these domains is strongly isoelectric point [16]. Therefore, it is believed that one of the
dependent on the pH value. A lower pH serves to change mechanisms by which pH triggers protein phase separation
the net charges and hydrophobicity of different domains as is the alteration of protein solubility (Fig. 3).
well as the interactions between these domains, leading to
significant structural reorganization. Thus, pH-mediated
diverse changes in α-Syn accelerate the maturation of phase pH changes act as messengers to transmit stress
separation and subsequent protein aggregation [149, 209]. signals
Similarly, a reduction in pH can enhance the intramolecular
interactions of the phase-separated SARS-CoV-2 N protein In the face of adverse conditions, intracellular pH might act
and lead to irregularly shaped assemblies with less liquid- as a messenger to signal changes in the environment, trig-
ity, in vitro [205]. Indeed, phase separation proteins that gering phase separation of proteins to promote cell fitness.
contain flexible LCDs are highly prone to forming patho- Upon heat shock, cells can integrate signals of different
genic aggregates. This could explain, to some extent, why temperatures and temperature-induced pH changes into a
hundreds of proteins are highly prone to forming aggregates unified response to provide a trigger for phase separation.
during aging. During aging or chronic pH stress, these phase For example, Pab1 undergoes LLPS autonomously through
separation proteins can transition into irreversible aggre- temperature-dependent structural changes under conditions
gates, which could then lead to persistent condensate for- of stressful temperatures [34].
mation, such as persistent SG formation, even after the stress However, how does a cell sense other stresses, such as
subsides. Persistent condensates typically exhibit solid-like starvation, to trigger LLPS to help cells survive diverse
properties; and as a consequence, other pathological changes adverse conditions? Previous studies have indicated that pro-
and neurodegenerative disorders occur [233]. teins and protein-associated condensates that undergo LLPS
under starvation conditions, such as Pub1, Gln1, Dhh1, and
pH changes affect protein solubility PAS, can also respond to low pH [47, 88, 192, 200, 201].
Considering that cytosolic pH is rapidly and reversibly reg-
In addition to engaging in the promiscuous interactions that ulated by glucose metabolism, the stress information per-
function in phase separation, macromolecules must reach ceived by these proteins is most likely transmitted through
a critical concentration threshold to start LLPS. Evidence pH. Evidence has shown that cytosolic pH is a second mes-
indicates that not all LCRs and IDRs function as autono- senger for glucose to mediate activation of the PKA pathway
mous modules that drive phase separation; instead, they through V-ATPase [43]. Therefore, a change in pH might
function as modifier sequences, regulating the solubility be an extremely sensitive readout of other changes in the
of phase-separating proteins and the material properties of environment, especially starvation, to induce protein LLPS
condensates [234]. Long-term evolutionary pressure has and cellular adaptive responses (Fig. 3).
tuned the solubility of Pub1 to be very close to the criti- In this way, pH is capable of playing diverse functional
cal threshold for phase separation. This not only endows roles in the regulation of LLPS, including by affecting pro-
Pub1 with a solubility that is conducive to growth but also tein–protein/RNA interactions, altering protein solubility,
enables Pub1 to quickly sense and respond to stress. In fact, and acting as a messenger to transmit stress signals.
13
13
biomolecular condensates. Cell 176(3):419–434. https://doi. 19. Orij R, Brul S, Smits GJ (2011) Intracellular pH is a tightly con-
org/10.1016/j.cell.2018.12.035 trolled signal in yeast. Biochim Biophys Acta 1810(10):933–944.
2. Hyman AA, Weber CA, Julicher F (2014) Liquid-liquid phase https://doi.org/10.1016/j.bbagen.2011.03.011
separation in biology. Annu Rev Cell Dev Biol 30:39–58. 20. Isom DG, Sridharan V, Baker R, Clement ST, Smalley DM,
https://doi.org/10.1146/annurev-cellbio-100913-013325 Dohlman HG (2013) Protons as second messenger regulators of
3. Boeynaems S, Alberti S, Fawzi NL, Mittag T, Polymenidou M, G protein signaling. Mol Cell 51(4):531–538. https://doi.org/10.
Rousseau F, Schymkowitz J, Shorter J, Wolozin B, Van Den 1016/j.molcel.2013.07.012
Bosch L, Tompa P, Fuxreiter M (2018) Protein phase separa- 21. Felle HH (2001) pH: signal and messenger in plant cells. Plant
tion: a new phase in cell biology. Trends Cell Biol 28(6):420– Biol 3(6):577–591. https://doi.org/10.1055/s-2001-19372
435. https://doi.org/10.1016/j.tcb.2018.02.004 22. Henderson KA, Hughes AL, Gottschling DE (2014) Mother-
4. Oldfield CJ, Dunker AK (2014) Intrinsically disordered pro- daughter asymmetry of pH underlies aging and rejuvenation in
teins and intrinsically disordered protein regions. Annu Rev yeast. Elife 3:e03504. https://doi.org/10.7554/eLife.03504
Biochem 83:553–584. https://doi.org/10.1146/annurev-bioch 23. Munder MC, Midtvedt D, Franzmann T, Nüske E, Otto O,
em-072711-164947 Herbig M, Ulbricht E, Müller P, Taubenberger A, Maharana S,
5. Gomes E, Shorter J (2019) The molecular language of mem- Malinovska L, Richter D, Guck J, Zaburdaev V, Alberti S (2016)
braneless organelles. J Biol Chem 294(18):7115–7127. https:// A pH-driven transition of the cytoplasm from a fluid- to a solid-
doi.org/10.1074/jbc.TM118.001192 like state promotes entry into dormancy. Elife 5:e09347. https://
6. Van Treeck B, Parker R (2018) Emerging roles for intermolecular doi.org/10.7554/eLife.09347
RNA-RNA interactions in RNP assemblies. Cell 174(4):791– 24. Ismail A, Takeda S, Nick P (2014) Life and death under salt
802. https://doi.org/10.1016/j.cell.2018.07.023 stress: same players, different timing? J Exp Bot 65(12):2963–
7. Van Treeck B, Protter DSW, Matheny T, Khong A, Link CD, 2979. https://doi.org/10.1093/jxb/eru159
Parker R (2018) RNA self-assembly contributes to stress granule 25. Obara M, Szeliga M, Albrecht J (2008) Regulation of pH in the
formation and defining the stress granule transcriptome. Proc mammalian central nervous system under normal and pathologi-
Natl Acad Sci USA 115(11):2734–2739. https://d oi.o rg/1 0.1 073/ cal conditions: facts and hypotheses. Neurochem Int 52(6):905–
pnas.1800038115 919. https://doi.org/10.1016/j.neuint.2007.10.015
8. Hofweber M, Dormann D (2019) Friend or foe-post-translational 26. Bright CM, Ellis D (1992) Intracellular pH changes induced by
modifications as regulators of phase separation and RNP granule hypoxia and anoxia in isolated sheep heart Purkinje fibres. Exp
dynamics. J Biol Chem 294(18):7137–7150. https://doi.org/10. Physiol 77(1):165–175. https://d oi.o rg/1 0.1 113/e xpphy siol.1 992.
1074/jbc.TM118.001189 sp003570
9. Luo YY, Wu JJ, Li YM (2021) Regulation of liquid-liquid phase 27. Diaz FE, Dantas E, Cabrera M, Benitez CA, Delpino MV, Duette
separation with focus on post-translational modifications. Chem G, Rubione J, Sanjuan N, Trevani AS, Geffner J (2016) Fever-
Commun (Camb) 57(98):13275–13287. https://doi.org/10.1039/ range hyperthermia improves the anti-apoptotic effect induced by
d1cc05266g low pH on human neutrophils promoting a proangiogenic profile.
10. Gao Y, Pei G, Li D, Li R, Shao Y, Zhang QC, Li P (2019) Cell Death Dis 7(10):e2437. https://doi.org/10.1038/cddis.2016.
Multivalent m6A motifs promote phase separation of YTHDF 337
proteins. Cell Res 29(9):767–769. https://d oi.o rg/1 0.1 038/ 28. Drummond IA, McClure SA, Poenie M, Tsien RY, Steinhardt RA
s41422-019-0210-3 (1986) Large changes in intracellular pH and calcium observed
11. Ries RJ, Zaccara S, Klein P, Olarerin-George A, Namkoong during heat shock are not responsible for the induction of heat
S, Pickering BF, Patil DP, Kwak H, Lee JH, Jaffrey SR (2019) shock proteins in Drosophila melanogaster. Mol Cell Biol
m6A enhances the phase separation potential of mRNA. Nature 6(5):1767–1775. https://doi.org/10.1128/mcb.6.5.1767-1775.
571(7765):424–428. https://d oi.o rg/1 0.1 038/s 41586-0 19-1 374-1 1986
12. Alberti S (2017) The wisdom of crowds: regulating cell func- 29. Ishizawa K (2014) Intracellular pH regulation of plant cells under
tion through condensed states of living matter. J Cell Sci anaerobic conditions. Plant Cell Monographs 21:59–74. https://
130(17):2789–2796. https://doi.org/10.1242/jcs.200295 doi.org/10.1007/978-3-7091-1254-0_4
13. Wang B, Zhang L, Dai T, Qin Z, Lu H, Zhang L, Zhou F (2021) 30. Weitzel G, Pilatus U, Rensing L (1987) The cytoplasmic pH,
Liquid-liquid phase separation in human health and diseases. ATP content and total protein synthesis rate during heat-shock
Signal Transduct Target Ther 6(1):290. https://doi.org/10.1038/ protein inducing treatments in yeast. Exp Cell Res 170(1):64–79.
s41392-021-00678-1 https://doi.org/10.1016/0014-4827(87)90117-0
14. Alberti S, Hyman AA (2016) Are aberrant phase transitions a 31. Webb BA, Chimenti M, Jacobson MP, Barber DL (2011) Dys-
driver of cellular aging? BioEssays 38(10):959–968. https://doi. regulated pH: a perfect storm for cancer progression. Nat Rev
org/10.1002/bies.201600042 Cancer 11(9):671–677. https://doi.org/10.1038/nrc3110
15. Ruff KM, Roberts S, Chilkoti A, Pappu RV (2018) Advances in 32. Roberts EL Jr, Sick TJ (1996) Aging impairs regulation of intra-
understanding stimulus-responsive phase behavior of intrinsi- cellular pH in rat hippocampal slices. Brain Res 735(2):339–342.
cally disordered protein polymers. J Mol Biol 430(23):4619– https://doi.org/10.1016/0006-8993(96)00925-0
4635. https://doi.org/10.1016/j.jmb.2018.06.031 33. Coote PJ, Cole MB, Jones MV (1991) Induction of increased
16. Ambadipudi S, Biernat J, Riedel D, Mandelkow E, Zweckstet- thermotolerance in Saccharomyces cerevisiae may be trig-
ter M (2017) Liquid-liquid phase separation of the microtubule- gered by a mechanism involving intracellular pH. J Gen
binding repeats of the Alzheimer-related protein Tau. Nat Com- Microbiol 137(7):1701–1708. https://doi.org/10.1099/00221
mun 8(1):275. https://doi.org/10.1038/s41467-017-00480-0 287-137-7-1701
17. Khan YM, East JM, Lee AG (1997) Effects of pH on phospho- 34. Riback JA, Katanski CD, Kear-Scott JL, Pilipenko EV, Rojek
rylation of the C a2+-ATPase of sarcoplasmic reticulum by inor- AE, Sosnick TR, Drummond DA (2017) Stress-triggered phase
ganic phosphate. Biochem J 321(Pt 3):671–676. https://doi.org/ separation is an adaptive, evolutionarily tuned response. Cell
10.1042/bj3210671 168(6):1028-1040.e19. https://doi.org/10.1016/j.cell.2017.02.
18. Mellman I (1992) The importance of being acid: the role of acidi- 027
fication in intracellular membrane traffic. J Exp Biol 172:39–45. 35. Triandafillou CG, Katanski CD, Dinner AR, Allan Drummond
https://doi.org/10.1242/jeb.172.1.39 D (2020) Transient intracellular acidification regulates the core
13
transcriptional heat shock response. Elife 9:e54880. https://doi. Appl Environ Microbiol 70(5):3176–3179. https://doi.org/10.
org/10.7554/eLife.54880 1128/AEM.70.5.3176-3179.2004
36. Pauli D, Arrigo A, Tissires A (1992) Heat shock response in 51. Csonka LN (1989) Physiological and genetic responses of bacte-
Drosophila. Experientia 48:623–629. https://doi.org/10.1007/ ria to osmotic stress. Microbiol Rev 53(1):121–147. https://doi.
BF02118306 org/10.1128/mmbr.53.1.121-147.1989
37. Zhong M, Kim SJ, Wu C (1999) Sensitivity of Drosophila heat 52. Dinnbier U, Limpinsel E, Schmid R, Bakker EP (1988) Transient
shock transcription factor to low pH. J Biol Chem 274(5):3135– accumulation of potassium glutamate and its replacement by tre-
3140. https://doi.org/10.1074/jbc.274.5.3135 halose during adaptation of growing cells of Escherichia coli
38. Aickin CC, Thomas RC (1977) An investigation of the ionic K-12 to elevated sodium chloride concentrations. Arch Microbiol
mechanism of intracellular pH regulation in mouse soleus muscle 150(4):348–357. https://doi.org/10.1007/BF00408306
fibres. J Physiol 273:295–316. https://doi.org/10.1113/jphysiol. 53. Castle AM, Macnab RM, Shulman RG (1986) Coupling between
1977.sp012095 the sodium and proton gradients in respiring Escherichia coli
39. Burdon RH, Cutmore CMM (1982) Human heat shock gene cells measured by 23Na and 31P nuclear magnetic resonance. J
expression and the modulation of plasma membrane Na+, K+- Biol Chem 261(17):7797–7806. https://doi.org/10.1016/s0021-
ATPase activity. FEBS Lett 140(1):45–48. https://doi.org/10. 9258(19)57471-3
1016/0014-5793(82)80517-6 54. Pintsch T, Satre M, Klein G, Martin JB, Schuster SC (2001)
40. Kiang JG, McKinney LC, Gallin EK (1990) Heat induces intra- Cytosolic acidification as a signal mediating hyperosmotic stress
cellular acidification in human A-431 cells: role of Na+-H+ responses in Dictyostelium discoideum. BMC Cell Biol 2(1):1–
exchange and metabolism. Am J Physiol 259(5 Pt 1):C727–C737. 15. https://doi.org/10.1186/1471-2121-2-9
https://doi.org/10.1152/ajpcell.1990.259.5.c727 55. Zischka H, Oehme F, Pintsch T, Ott A, Keller H, Kellermann
41. Lamarche S, Chretien P, Landry J (1985) Inhibition of the heat J, Schuster SC (1999) Rearrangement of cortex proteins consti-
shock response and synthesis of glucose-regulated proteins in tutes an osmoprotective mechanism in Dictyostelium. EMBO J
Ca2+-deprived rat hepatoma cells. Biochem Biophys Res Com- 18(15):4241–4249. https://doi.org/10.1093/emboj/18.15.4241
mun 131(2):868–876. https://doi.org/10.1016/0006-291x(85) 56. Oyama M, Kubota K (1997) H + secretion induced by hyper-
91320-8 tonic stress in the cellular slime mold Dictyostelium discoideum.
42. Lepock JR, Cheng KH, Al-Qysi H, Kruuv J (1983) Thermotropic J Biochem 122(1):64–70. https://d oi.o rg/1 0.1 093/o xford journ als.
lipid and protein transitions in chinese hamster lung cell mem- jbchem.a021741
branes: relationship to hyperthermic cell killing. Can J Biochem 57. Bracey D, Holyoak CD, Nebe-Von Caron G, Coote PJ (1998)
Cell Biol 61(6):421–427. https://doi.org/10.1139/o83-057 Determination of the intracellular pH (pHi) of growing cells of
43. Dechant R, Binda M, Lee SS, Pelet S, Winderickx J, Peter Saccharomyces cerevisiae: the effect of reduced-expression of
M (2010) Cytosolic pH is a second messenger for glucose the membrane H +-ATPase. J Microbiol Methods 31(3):113–125.
and regulates the PKA pathway through V-ATPase. EMBO J https://doi.org/10.1016/S0167-7012(97)00095-X
29(15):2515–2526. https://doi.org/10.1038/emboj.2010.138 58. Cole MB, Keenan MHJ (1987) Effects of weak acids and external
44. Franzmann TM, Jahnel M, Pozniakovsky A, Mahamid J, pH on the intracellular pH of Zygosaccharomyces bailii, and its
Holehouse AS, Nüske E, Richter D, Baumeister W, Grill implications in weak-acid resistance. Yeast 3(1):23–32. https://
SW, Pappu RV, Hyman AA, Alberti S (2018) Phase separa- doi.org/10.1002/yea.320030105
tion of a yeast prion protein promotes cellular fitness. Science 59. Salmond CV, Kroll RG, Booth IR (1984) The effect of food
359(6371):eaao5654. https://doi.org/10.1126/science.aao5654 preservatives on pH homeostasis in Escherichia coli. J Gen
45. Orij R, Urbanus ML, Vizeacoumar FJ, Giaever G, Boone C, Microbiol 130(11):2845–2850. https://doi.org/10.1099/00221
Nislow C, Brul S, Smits GJ (2012) Genome-wide analysis of 287-130-11-2845
intracellular pH reveals quantitative control of cell division rate 60. Yao H, Haddad GG (2004) Calcium and pH homeostasis in neu-
by pHc in Saccharomyces cerevisiae. Genome Biol 13(9):R80. rons during hypoxia and ischemia. Cell Calcium 36(3–4):247–
https://doi.org/10.1186/gb-2012-13-9-r80 255. https://doi.org/10.1016/j.ceca.2004.02.013
46. Orij R, Postmus J, Beek AT, Brul S, Smits GJ (2009) In vivo 61. Roberts JK, Callis J, Wemmer D, Walbot V, Jardetzky O (1984)
measurement of cytosolic and mitochondrial pH using a pH- Mechanisms of cytoplasmic pH regulation in hypoxic maize root
sensitive GFP derivative in Saccharomyces cerevisiae reveals tips and its role in survival under hypoxia. Proc Natl Acad Sci
a relation between intracellular pH and growth. Microbiol- USA 81(11):3379–3383. https://doi.org/10.1073/pnas.81.11.
ogy (Reading) 155(1):268–278. https://doi.org/10.1099/mic.0. 3379
022038-0 62. Davies DD (1980) Anaerobic metabolism and the production of
47. Petrovska I, Nüske E, Munder MC, Kulasegaran G, Malinovska organic acids. Metab Resp 2:581–611. https://doi.org/10.1016/
L, Kroschwald S, Richter D, Fahmy K, Gibson K, Verbavatz JM, b978-0-12-675402-5.50020-9
Alberti S (2014) Filament formation by metabolic enzymes is a 63. Leão C, Van Uden N (1984) Effects of ethanol and other alkanols
specific adaptation to an advanced state of cellular starvation. on passive proton influx in the yeast Saccharomyces cerevisiae.
Elife 2014(3):e02409. https://doi.org/10.7554/eLife.02409 Biochim Biophys Acta 774(1):43–48. https://doi.org/10.1016/
48. Morisawa M, Steinhardt RA (1982) Changes in intracellular pH 0005-2736(84)90272-4
of Physarum plasmodium during the cell cycle and in response 64. Li GC, Shiu EC, Hahn GM (1980) Similarities in cellular inacti-
to starvation. Exp Cell Res 140(2):341–351. https://doi.org/10. vation by hyperthermia or by ethanol. Radiat Res 82(2):257–268.
1016/0014-4827(82)90123-9 https://doi.org/10.2307/3575377
49. Chakraborty S, Winardhi RS, Morgan LK, Yan J, Kenney LJ 65. He DY, Yazaki Y, Nishizawa Y, Takai R, Yamada K, Sakano K,
(2017) Non-canonical activation of OmpR drives acid and Shibuya N, Minami E (1998) Gene activation by cytoplasmic
osmotic stress responses in single bacterial cells. Nat Commun acidification in suspension-cultured rice cells in response to the
8(1):1587. https://doi.org/10.1038/s41467-017-02030-0 potent elicitor, N-acetylchitoheptaose. Mol Plant Microbe Inter-
50. Fang W, Siegumfeldt H, Budde BB, Jakobsen M (2004) Osmotic act 11(12):1167–1174. https://doi.org/10.1094/MPMI.1998.11.
stress leads to decreased intracellular pH of Listeria monocy- 12.1167
togenes as determined by fluorescence ratio-imaging microscopy. 66. Hansen UP, Moldaenke C, Tabrizi H, Ramm D (1993) The
effect of transthylakoid proton uptake on cytosolic pH and the
13
imbalance of ATP and NAPDH/H+ production as measured by 82. Nivon M, Richet E, Codogno P, Arrigo AP, Kretz-Remy C (2009)
CO2- and light-induced depolarisation of the plasmalemma. Plant Autophagy activation by NFkappaB is essential for cell survival
Cell Physiol 34(5):681–695. https://doi.org/10.1093/oxfordjour after heat shock. Autophagy 5(6):766–783. https://doi.org/10.
nals.pcp.a078471 4161/auto.8788
67. Hughes AL, Gottschling DE (2012) An early age increase in 83. Weitzel G, Pilatus U, Rensing L (1985) Similar dose response
vacuolar pH limits mitochondrial function and lifespan in yeast. of heat shock protein synthesis and intracellular pH change in
Nature 492(7428):261–265. https://d oi.o rg/1 0.1 038/n ature 11654 yeast. Exp Cell Res 159(1):252–256. https://doi.org/10.1016/
68. van Schalkwyk DA, Saliba KJ, Biagini GA, Bray PG, Kirk K s0014-4827(85)80054-9
(2013) Loss of pH control in Plasmodium falciparum parasites 84. Tombaugh GC, Sapolsky RM (1993) Evolving concepts about
subjected to oxidative stress. PLoS ONE 8(3):e58933. https:// the role of acidosis in ischemic neuropathology. J Neurochem
doi.org/10.1371/journal.pone.0058933 61(3):793–803. https://doi.org/10.1111/j.1471-4159.1993.tb035
69. Wang Y, Floor E (1998) Hydrogen peroxide inhibits the vacuolar 89.x
H+-ATPase in brain synaptic vesicles at micromolar concentra- 85. Joyner RP, Tang JH, Helenius J, Dultz E, Brune C, Holt LJ, Huet
tions. J Neurochem 70(2):646–652. https://doi.org/10.1046/j. S, Muller DJ, Weis K (2016) A glucose-starvation response regu-
1471-4159.1998.70020646.x lates the diffusion of macromolecules. Elife 5:e09376. https://d oi.
70. Vihervaara A, Sistonen L (2014) HSF1 at a glance. J Cell Sci org/10.7554/eLife.09376
127(Pt 2):261–266. https://doi.org/10.1242/jcs.132605 86. Jayaraj GG, Hipp MS, Hartl FU (2020) Functional modules
71. Cabrera M, Boronat S, Marte L, Vega M, Perez P, Ayte J, Hidalgo of the proteostasis network. Cold Spring Harb Perspect Biol
E (2020) Chaperone-facilitated aggregation of thermo-sensitive 12(1):a033951. https://doi.org/10.1101/cshperspect.a033951
proteins shields them from degradation during heat stress. Cell 87. Iserman C, Desroches Altamirano C, Jegers C, Friedrich U, Zarin
Rep 30(7):2430-2443.e4. https://doi.org/10.1016/j.celrep.2020. T, Fritsch AW, Mittasch M, Domingues A, Hersemann L, Jahnel
01.077 M, Richter D, Guenther UP, Hentze MW, Moses AM, Hyman
72. Kim JH, Park SJ, Kim TS, Park HJ, Park J, Kim BK, Kim GR, AA, Kramer G, Kreysing M, Franzmann TM, Alberti S (2020)
Kim JM, Huang SM, Chae JI, Park CK, Lee DS (2013) Tes- Condensation of Ded1p promotes a translational switch from
ticular hyperthermia induces unfolded protein response signal- housekeeping to stress protein production. Cell 181(4):818-831.
ing activation in spermatocyte. Biochem Biophys Res Commun e19. https://doi.org/10.1016/j.cell.2020.04.009
434(4):861–866. https://doi.org/10.1016/j.bbrc.2013.04.032 88. Kroschwald S, Munder MC, Maharana S, Franzmann TM, Rich-
73. Mizusawa M, Sharmin MM, Yonekura S (2019) Mild heat stress ter D, Ruer M, Hyman AA, Alberti S (2018) Different material
induces transcription of the beta-casein gene via unfolded protein states of Pub1 condensates define distinct modes of stress adapta-
response-activated XBP1 signaling in undifferentiated mammary tion and recovery. Cell Rep 23(11):3327–3339. https://doi.org/
epithelial cells. Anim Sci J 90(8):1026–1032. https://doi.org/10. 10.1016/j.celrep.2018.05.041
1111/asj.13246 89. Yang P, Mathieu C, Kolaitis RM, Zhang P, Messing J, Yurtsever
74. Maxwell BA, Gwon Y, Mishra A, Peng J, Nakamura H, U, Yang Z, Wu J, Li Y, Pan Q, Yu J, Martin EW, Mittag T, Kim
Zhang K, Kim HJ, Taylor JP (2021) Ubiquitination is essen- HJ, Taylor JP (2020) G3BP1 is a tunable switch that triggers
tial for recovery of cellular activities after heat shock. Science phase separation to assemble stress granules. Cell 181(2):325-
372(6549):eabc3593. https://doi.org/10.1126/science.abc3593 345.e28. https://doi.org/10.1016/j.cell.2020.03.046
75. Carlson N, Rogers S, Rechsteiner M (1987) Microinjection of 90. Piper PW (1993) Molecular events associated with acquisition
ubiquitin: changes in protein degradation in HeLa cells subjected of heat tolerance by the yeast Saccharomyces cerevisiae. FEMS
to heat-shock. J Cell Biol 104(3):547–555. https://doi.org/10. Microbiol Rev 11(4):339–355. https://doi.org/10.1111/j.1574-
1083/jcb.104.3.547 6976.1993.tb00005.x
76. Medicherla B, Goldberg AL (2008) Heat shock and oxygen radi- 91. Panaretou B, Piper PW (1990) Plasma-membrane ATPase action
cals stimulate ubiquitin-dependent degradation mainly of newly affects several stress tolerances of Saccharomyces cerevisiae and
synthesized proteins. J Cell Biol 182(4):663–673. https://d oi.o rg/ Schizosaccharomyces pombe as well as the extent and duration
10.1083/jcb.200803022 of the heat shock response. J Gen Microbiol 136:1763–1770.
77. Parag HA, Raboy B, Kulka RG (1987) Effect of heat shock on https://doi.org/10.1099/00221287-136-9-1763
protein degradation in mammalian cells: involvement of the 92. Morsomme P, Slayman CW, Goffeau A (2000) Mutagenic study
ubiquitin system. EMBO J 6(1):55–61. https://doi.org/10.1002/j. of the structure, function and biogenesis of the yeast plasma
1460-2075.1987.tb04718.x membrane H+-ATPase. Biochim Biophys Acta 1469(3):133–157.
78. Dokladny K, Zuhl MN, Mandell M, Bhattacharya D, Schnei- https://doi.org/10.1016/s0304-4157(00)00015-0
der S, Deretic V, Moseley PL (2013) Regulatory coordination 93. Perlin DS, San Francisco MJ, Slayman CW, Rosen BP (1986) H+/
between two major intracellular homeostatic systems: heat shock ATP stoichiometry of proton pumps from Neurospora crassa and
response and autophagy. J Biol Chem 288(21):14959–14972. Escherichia coli. Arch Biochem Biophys 248(1):53–61. https://
https://doi.org/10.1074/jbc.M113.462408 doi.org/10.1016/0003-9861(86)90400-5
79. Zhao Y, Gong S, Shunmei E, Zou J (2009) Induction of macro- 94. Lecchi S, Nelson CJ, Allen KE, Swaney DL, Thompson KL,
autophagy by heat. Mol Biol Rep 36(8):2323–2327. https://doi. Coon JJ, Sussman MR, Slayman CW (2007) Tandem phos-
org/10.1007/s11033-009-9451-4 phorylation of Ser-911 and Thr-912 at the C terminus of yeast
80. Hsu SF, Chao CM, Huang WT, Lin MT, Cheng BC (2013) Atten- plasma membrane H+-ATPase leads to glucose-dependent activa-
uating heat-induced cellular autophagy, apoptosis and damage in tion. J Biol Chem 282(49):35471–35481. https://d oi.o rg/1 0.1 074/
H9c2 cardiomyocytes by pre-inducing HSP70 with heat shock jbc.M706094200
preconditioning. Int J Hyperthermia 29(3):239–247. https://doi. 95. Martinez-Munoz GA, Kane P (2008) Vacuolar and plasma mem-
org/10.3109/02656736.2013.777853 brane proton pumps collaborate to achieve cytosolic pH homeo-
81. Zhang M, Jiang M, Bi Y, Zhu H, Zhou Z, Sha J (2012) Autophagy stasis in yeast. J Biol Chem 283(29):20309–20319. https://doi.
and apoptosis act as partners to induce germ cell death after heat org/10.1074/jbc.M710470200
stress in mice. PLoS ONE 7(7):e41412. https://doi.org/10.1371/ 96. Perzov N, Nelson H, Nelson N (2000) Altered distribution of
journal.pone.0041412 the yeast plasma membrane H +-ATPase as a feature of vacuolar
13
13
neurons. Front Physiol 5:43. https://doi.org/10.3389/fphys.2014. amyotrophic lateral sclerosis. Science 314(5796):130–133.
00043 https://doi.org/10.1126/science.1134108
131. Raffin CN, Sick TJ, Rosenthal M (1988) Inhibition of glycolysis 145. Neumann M, Bentmann E, Dormann D, Jawaid A, DeJesus-
alters potassium ion transport and mitochondrial redox activity Hernandez M, Ansorge O, Roeber S, Kretzschmar HA, Munoz
in rat brain. J Cereb Blood Flow Metab 8(6):857–865. https:// DG, Kusaka H, Yokota O, Ang LC, Bilbao J, Rademakers R,
doi.org/10.1038/jcbfm.1988.143 Haass C, Mackenzie IR (2011) FET proteins TAF15 and EWS
132. Bonnet U, Bingmann D, Wiltfang J, Scherbaum N, Wiemann M are selective markers that distinguish FTLD with FUS pathology
(2010) Modulatory effects of neuropsychopharmaca on intra- from amyotrophic lateral sclerosis with FUS mutations. Brain
cellular pH of hippocampal neurones in vitro. Br J Pharmacol 134(Pt 9):2595–2609. https://doi.org/10.1093/brain/awr201
159(2):474–483. https://doi.org/10.1111/j.1476-5381.2009. 146. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes
00540.x R, Goedert M (1997) Alpha-synuclein in Lewy bodies. Nature
133. Baram TZ, Eghbal-Ahmadi M, Bender RA (2002) Is neuronal 388(6645):839–840. https://doi.org/10.1038/42166
death required for seizure-induced epileptogenesis in the imma- 147. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS,
ture brain? Prog Brain Res 135:365–375. https://d oi.o rg/1 0.1 016/ Wisniewski HM (1986) Microtubule-associated protein tau. A
S0079-6123(02)35033-7 component of Alzheimer paired helical filaments. J Biol Chem
134. Roberts EL Jr, Rosenthal M, Sick TJ (1990) Age-related modi- 261(13):6084–6089. https://doi.org/10.1016/S0021-9258(17)
fications of potassium homeostasis and synaptic transmission 38495-8
during and after anoxia in rat hippocampal slices. Brain Res 148. Zbinden A, Perez-Berlanga M, De Rossi P, Polymenidou M
514(1):111–118. https://d oi.o rg/1 0.1 016/0 006-8 993(90)9 0441-d (2020) Phase separation and neurodegenerative diseases: a dis-
135. Yao H, Sadoshima S, Ooboshi H, Sato Y, Uchimura H, turbance in the force. Dev Cell 55(1):45–68. https://doi.org/10.
Fujishima M (1991) Age-related vulnerability to cer- 1016/j.devcel.2020.09.014
ebral ischemia in spontaneously hypertensive rats. Stroke 149. Ray S, Singh N, Kumar R, Patel K, Pandey S, Datta D, Mahato
22(11):1414–1418. https://doi.org/10.1161/01.str.22.11.1414 J, Panigrahi R, Navalkar A, Mehra S, Gadhe L, Chatterjee D,
136. Colacurcio DJ, Nixon RA (2016) Disorders of lysosomal acid- Sawner AS, Maiti S, Bhatia S, Gerez JA, Chowdhury A, Kumar
ification-the emerging role of v-ATPase in aging and neurode- A, Padinhateeri R, Riek R, Krishnamoorthy G, Maji SK (2020)
generative disease. Ageing Res Rev 32:75–88. https://doi.org/ α-Synuclein aggregation nucleates through liquid–liquid phase
10.1016/j.arr.2016.05.004 separation. Nat Chem 12(8):705–716. https://doi.org/10.1038/
137. Ruckenstuhl C, Netzberger C, Entfellner I, Carmona-Gut- s41557-020-0465-9
ierrez D, Kickenweiz T, Stekovic S, Gleixner C, Schmid C, 150. Bouchard JJ, Otero JH, Scott DC, Szulc E, Martin EW, Sabri
Klug L, Sorgo AG, Eisenberg T, Buttner S, Marino G, Koz- N, Granata D, Marzahn MR, Lindorff-Larsen K, Salvatella X,
iel R, Jansen-Durr P, Frohlich KU, Kroemer G, Madeo F Schulman BA, Mittag T (2018) Cancer mutations of the tumor
(2014) Lifespan extension by methionine restriction requires suppressor SPOP disrupt the formation of active, phase-sepa-
autophagy-dependent vacuolar acidification. PLoS Genet rated compartments. Mol Cell 72(1):19-36.e8. https://doi.org/
10(5):e1004347. https://doi.org/10.1371/journal.pgen.1004347 10.1016/j.molcel.2018.08.027
138. Molin M, Demir AB (2014) Linking peroxiredoxin and vac- 151. Kilic S, Lezaja A, Gatti M, Bianco E, Michelena J, Imhof R,
uolar-ATPase functions in calorie restriction-mediated life Altmeyer M (2019) Phase separation of 53BP1 determines
span extension. Int J Cell Biol 2014:913071. https://doi.org/ liquid-like behavior of DNA repair compartments. EMBO J
10.1155/2014/913071 38(16):e101379. https://doi.org/10.15252/embj.2018101379
139. Di Domenico F, Perluigi M, Butterfield DA, Cornelius C, Cala- 152. Altmeyer M, Neelsen KJ, Teloni F, Pozdnyakova I, Pellegrino
brese V (2010) Oxidative damage in rat brain during aging: S, Grofte M, Rask MD, Streicher W, Jungmichel S, Nielsen
interplay between energy and metabolic key target proteins. ML, Lukas J (2015) Liquid demixing of intrinsically disordered
Neurochem Res 35(12):2184–2192. https://doi.org/10.1007/ proteins is seeded by poly(ADP-ribose). Nat Commun 6:8088.
s11064-010-0295-z https://doi.org/10.1038/ncomms9088
140. Fujisawa Y, Kato K, Giulivi C (2009) Nitration of tyrosine 153. Boulay G, Sandoval GJ, Riggi N, Iyer S, Buisson R, Naigles B,
residues 368 and 345 in the β-subunit elicits FoF 1-ATPase Awad ME, Rengarajan S, Volorio A, McBride MJ, Broye LC,
activity loss. Biochem J 423(2):219–231. https://doi.org/10. Zou L, Stamenkovic I, Kadoch C, Rivera MN (2017) Cancer-
1042/BJ20090594 specific retargeting of BAF complexes by a prion-like domain.
141. Haynes V, Traaseth NJ, Elfering S, Fujisawa Y, Giulivi C Cell 171(1):163-178.e19. https://doi.org/10.1016/j.cell.2017.07.
(2010) Nitration of specific tyrosines in FoF1 ATP synthase 036
and activity loss in aging. Am J Physiol Endocrinol Metab 154. Zamudio AV, Dall’Agnese A, Henninger JE, Manteiga JC,
298(5):E978–E987. https://d oi.o rg/1 0.1 152/ajpend o.0 0739. Afeyan LK, Hannett NM, Coffey EL, Li CH, Oksuz O, Sabari
2009 BR, Boija A, Klein IA, Hawken SW, Spille JH, Decker TM,
142. Ginsberg SD, Alldred MJ, Counts SE, Cataldo AM, Neve RL, Cisse II, Abraham BJ, Lee TI, Taatjes DJ, Schuijers J, Young
Jiang Y, Wuu J, Chao MV, Mufson EJ, Nixon RA, Che S (2010) RA (2019) Mediator condensates localize signaling factors to
Microarray analysis of hippocampal CA1 neurons implicates key cell identity genes. Mol Cell 76(5):753-766.e6. https://doi.
early endosomal dysfunction during Alzheimer’s disease pro- org/10.1016/j.molcel.2019.08.016
gression. Biol Psychiatry 68(10):885–893. https://doi.org/10. 155. Lu Y, Wu T, Gutman O, Lu H, Zhou Q, Henis YI, Luo K (2020)
1016/j.biopsych.2010.05.030 Phase separation of TAZ compartmentalizes the transcription
143. Taylor RC, Dillin A (2011) Aging as an event of proteostasis machinery to promote gene expression. Nat Cell Biol 22(4):453–
collapse. Cold Spring Harb Perspect Biol 3(5):a004440. https:// 464. https://doi.org/10.1038/s41556-020-0485-0
doi.org/10.1101/cshperspect.a004440 156. Arimoto K, Fukuda H, Imajoh-Ohmi S, Saito H, Takekawa
144. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi M (2008) Formation of stress granules inhibits apoptosis by
MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, suppressing stress-responsive MAPK pathways. Nat Cell Biol
McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman 10(11):1324–1332. https://doi.org/10.1038/ncb1791
H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM (2006) 157. Thedieck K, Holzwarth B, Prentzell MT, Boehlke C, Klasener
Ubiquitinated TDP-43 in frontotemporal lobar degeneration and K, Ruf S, Sonntag AG, Maerz L, Grellscheid SN, Kremmer E,
13
Nitschke R, Kuehn EW, Jonker JW, Groen AK, Reth M, Hall 174. Roos A, Boron WF (1981) Intracellular pH. Physiol Rev
MN, Baumeister R (2013) Inhibition of mTORC1 by astrin 61(2):296–434. https://doi.org/10.1152/physrev.1981.61.2.296
and stress granules prevents apoptosis in cancer cells. Cell 175. Sakano K (2001) Metabolic regulation of pH in plant cells: role
154(4):859–874. https://doi.org/10.1016/j.cell.2013.07.031 of cytoplasmic pH in defense reaction and secondary metabo-
158. Hsu KS, Kao HY (2018) PML: Regulation and multifaceted lism. Int Rev Cytol 206:1–44. https://doi.org/10.1016/s0074-
function beyond tumor suppression. Cell Biosci 8:5. https://doi. 7696(01)06018-1
org/10.1186/s13578-018-0204-8 176. Davies DD (1986) The fine control of cytosolic pH. Physiol Plant
159. Adriaens C, Standaert L, Barra J, Latil M, Verfaillie A, Kalev 67(4):702–706. https://doi.org/10.1111/j.1399-3054.1986.tb050
P, Boeckx B, Wijnhoven PW, Radaelli E, Vermi W, Leucci E, 81.x
Lapouge G, Beck B, van den Oord J, Nakagawa S, Hirose T, 177. Hurth MA, Suh SJ, Kretzschmar T, Geis T, Bregante M, Gambale
Sablina AA, Lambrechts D, Aerts S, Blanpain C, Marine JC F, Martinoia E, Neuhaus HE (2005) Impaired pH homeostasis in
(2016) p53 induces formation of NEAT1 lncRNA-containing Arabidopsis lacking the vacuolar dicarboxylate transporter and
paraspeckles that modulate replication stress response and che- analysis of carboxylic acid transport across the tonoplast. Plant
mosensitivity. Nat Med 22(8):861–868. https://doi.org/10.1038/ Physiol 137(3):901–910. https://doi.org/10.1104/pp.104.058453
nm.4135 178. Sigler K, Kotyk A, Knotkova A, Opekarova M (1981) Processes
160. Audas TE, Audas DE, Jacob MD, Ho JJ, Khacho M, Wang M, involved in the creation of buffering capacity and in substrate-
Perera JK, Gardiner C, Bennett CA, Head T, Kryvenko ON, induced proton extrusion in the yeast Saccharomyces cerevisiae.
Jorda M, Daunert S, Malhotra A, Trinkle-Mulcahy L, Gon- Biochim Biophys Acta 643(3):583–592. https://doi.org/10.1016/
zalgo ML, Lee S (2016) Adaptation to stressors by systemic 0005-2736(81)90354-0
protein amyloidogenesis. Dev Cell 39(2):155–168. https://doi. 179. Michenkova M, Taki S, Blosser MC, Hwang HJ, Kowatz T, Moss
org/10.1016/j.devcel.2016.09.002 FJ, Occhipinti R, Qin X, Sen S, Shinn E, Wang D, Zeise BS,
161. Lane DP (1992) Cancer. p53, guardian of the genome. Nature Zhao P, Malmstadt N, Vahedi-Faridi A, Tajkhorshid E, Boron
358(6381):15–16. https://doi.org/10.1038/358015a0 WF (2021) Carbon dioxide transport across membranes. Inter-
162. Levine AJ (1997) p53, the cellular gatekeeper for growth and face Focus 11(2):20200090. https://doi.org/10.1098/rsfs.2020.
division. Cell 88(3):323–331. https://doi.org/10.1016/s0092- 0090
8674(00)81871-1 180. Kane PM (2006) The where, when, and how of organelle acidi-
163. Levine AJ, Momand J, Finlay CA (1991) The p53 tumour sup- fication by the yeast vacuolar H +-ATPase. Microbiol Mol Biol
pressor gene. Nature 351(6326):453–456. https://doi.org/10. Rev 70(1):177–191. https://doi.org/10.1128/MMBR.70.1.177-
1038/351453a0 191.2006
164. Vousden KH, Lu X (2002) Live or let die: the cell’s response 181. Kobayashi H, Suzuki T, Unemoto T (1986) Streptococcal cyto-
to p53. Nat Rev Cancer 2(8):594–604. https://doi.org/10.1038/ plasmic pH is regulated by changes in amount and activity of
nrc864 a proton-translocating ATPase. J Biol Chem 261(2):627–630.
165. Freed-Pastor WA, Prives C (2012) Mutant p53: one name, https://doi.org/10.1016/s0021-9258(17)36138-0
many proteins. Genes Dev 26(12):1268–1286. https://doi.org/ 182. Kobayashi H (2003) Computer simulation of cytoplasmic pH
10.1101/gad.190678.112 regulation mediated by the F-type H+-ATPase. Biochim Biophys
166. Silva JL, De Moura Gallo CV, Costa DC, Rangel LP (2014) Acta 1607(2–3):211–216. https://d oi.o rg/1 0.1 016/j.b babio.2 003.
Prion-like aggregation of mutant p53 in cancer. Trends Bio- 10.001
chem Sci 39(6):260–267. https://doi.org/10.1016/j.tibs.2014. 183. Cordat E, Casey JR (2009) Bicarbonate transport in cell physiol-
04.001 ogy and disease. Biochem J 417(2):423–439. https://doi.org/10.
167. Petronilho EC, Pedrote MM, Marques MA, Passos YM, Mota 1042/BJ20081634
MF, Jakobus B, de Sousa GDS, Pereira da Costa F, Felix AL, 184. Gotz R, Gnann A, Zimmermann FK (1999) Deletion of the
Ferretti GDS, Almeida FP, Cordeiro Y, Vieira T, de Oliveira carbonic anhydrase-like gene NCE103 of the yeast Saccha-
GAP, Silva JL (2021) Phase separation of p53 precedes aggrega- romyces cerevisiae causes an oxygen-sensitive growth defect.
tion and is affected by oncogenic mutations and ligands. Chem Yeast 15(10A):855–864. https://doi.org/10.1002/(SICI)1097-
Sci 12(21):7334–7349. https://doi.org/10.1039/d1sc01739j 0061(199907)15:10A%3c855::AID-YEA425%3e3.0.CO;2-C
168. Ackerman D, Simon MC (2014) Hypoxia, lipids, and cancer: 185. Gawenis LR, Ledoussal C, Judd LM, Prasad V, Alper SL, Stu-
surviving the harsh tumor microenvironment. Trends Cell Biol art-Tilley A, Woo AL, Grisham C, Sanford LP, Doetschman T,
24(8):472–478. https://doi.org/10.1016/j.tcb.2014.06.001 Miller ML, Shull GE (2004) Mice with a targeted disruption of
169. Gillies RJ, Raghunand N, Karczmar GS, Bhujwalla ZM (2002) the AE2 C l-/HCO3- exchanger are achlorhydric. J Biol Chem
MRI of the tumor microenvironment. J Magn Reson Imaging 279(29):30531–30539. https://d oi.o rg/1 0.1 074/j bc.M
40377 9200
16(4):430–450. https://doi.org/10.1002/jmri.10181 186. Guenther UP, Weinberg DE, Zubradt MM, Tedeschi FA, Stawicki
170. Stuwe L, Muller M, Fabian A, Waning J, Mally S, Noel J, Schwab BN, Zagore LL, Brar GA, Licatalosi DD, Bartel DP, Weissman
A, Stock C (2007) pH dependence of melanoma cell migration: JS, Jankowsky E (2018) The helicase Ded1p controls use of
protons extruded by NHE1 dominate protons of the bulk solution. near-cognate translation initiation codons in 5′ UTRs. Nature
J Physiol 585(Pt 2):351–360. https://doi.org/10.1113/jphysiol. 559(7712):130–134. https://d oi.o rg/1 0.1 038/s 41586-0 18-0 258-0
2007.145185 187. Sen ND, Zhou F, Ingolia NT, Hinnebusch AG (2015) Genome-
171. Kane PM (2016) Proton transport and pH control in fungi. wide analysis of translational efficiency reveals distinct but over-
Adv Exp Med Biol 892:33–68. https:// d oi. o rg/ 1 0. 1 007/ lapping functions of yeast DEAD-box RNA helicases Ded1 and
978-3-319-25304-6_3 eIF4A. Genome Res 25(8):1196–1205. https://doi.org/10.1101/
172. Casey JR, Grinstein S, Orlowski J (2010) Sensors and regulators gr.191601.115
of intracellular pH. Nat Rev Mol Cell Biol 11(1):50–61. https:// 188. Hilliker A, Gao Z, Jankowsky E, Parker R (2011) The DEAD-box
doi.org/10.1038/nrm2820 protein Ded1 modulates translation by the formation and reso-
173. Pittman JK (2012) Multiple transport pathways for mediat- lution of an eIF4F-mRNA complex. Mol Cell 43(6):962–972.
ing intracellular pH homeostasis: the contribution of H+/ion https://doi.org/10.1016/j.molcel.2011.08.008
exchangers. Front Plant Sci 3:11. https://doi.org/10.3389/fpls.
2012.00011
13
189. Kroschwald S, Alberti S (2017) Gel or die: phase separation as a Alberti S, Franzmann TM (2020) RNA-induced conformational
survival strategy. Cell 168(6):947–948. https://d oi.o rg/1 0.1 016/j. switching and clustering of G3BP drive stress granule assembly
cell.2017.02.029 by condensation. Cell 181(2):346-361.e17. https://doi.org/10.
190. Yao G, Chiang Y-C, Zhang C, Lee DJ, Laue TM, Denis CL 1016/j.cell.2020.03.049
(2007) PAB1 self-association precludes its binding to poly(A), 204. Luo L, Li Z, Zhao T, Ju X, Ma P, Jin B, Zhou Y, He S, Huang J,
thereby accelerating CCR4 deadenylation in vivo. Mol Cell Biol Xu X, Zou Y, Li P, Liang A, Liu J, Chi T, Huang X, Ding Q, Jin
27(17):6243–6253. https://doi.org/10.1128/mcb.00734-07 Z, Huang C, Zhang Y (2021) SARS-CoV-2 nucleocapsid protein
191. Alberti S, Halfmann R, King O, Kapila A, Lindquist S (2009) phase separates with G3BPs to disassemble stress granules and
A systematic survey identifies prions and illuminates sequence facilitate viral production. Sci Bull (Beijing) 66(12):1194–1204.
features of prionogenic proteins. Cell 137(1):146–158. https:// https://doi.org/10.1016/j.scib.2021.01.013
doi.org/10.1016/j.cell.2009.02.044 205. Perdikari TM, Murthy AC, Ryan VH, Watters S, Naik MT, Fawzi
192. Hondele M, Sachdev R, Heinrich S, Wang J, Vallotton P, Fon- NL (2020) SARS-CoV-2 nucleocapsid protein phase-separates
toura BMA, Weis K (2019) DEAD-box ATPases are global regu- with RNA and with human hnRNPs. EMBO J 39(24):e106478.
lators of phase-separated organelles. Nature 573(7772):144–148. https://doi.org/10.15252/embj.2020106478
https://doi.org/10.1038/s41586-019-1502-y 206. You J, Dove BK, Enjuanes L, DeDiego ML, Alvarez E, Howell
193. Nott TJ, Petsalaki E, Farber P, Jervis D, Fussner E, Plochowietz G, Heinen P, Zambon M, Hiscox JA (2005) Subcellular localiza-
A, Craggs TD, Bazett-Jones DP, Pawson T, Forman-Kay JD, tion of the severe acute respiratory syndrome coronavirus nucle-
Baldwin AJ (2015) Phase transition of a disordered nuage protein ocapsid protein. J Gen Virol 86(Pt 12):3303–3310. https://doi.
generates environmentally responsive membraneless organelles. org/10.1099/vir.0.81076-0
Mol Cell 57(5):936–947. https://doi.org/10.1016/j.molcel.2015. 207. Mehra S, Sahay S, Maji SK (2019) α-Synuclein misfolding and
01.013 aggregation: implications in Parkinson’s disease pathogenesis.
194. Linder P, Jankowsky E (2011) From unwinding to clamping— Biochim Biophys Acta Proteins Proteom 1867(10):890–908.
the DEAD box RNA helicase family. Nat Rev Mol Cell Biol https://doi.org/10.1016/j.bbapap.2019.03.001
12(8):505–516. https://doi.org/10.1038/nrm3154 208. Lashuel HA, Overk CR, Oueslati A, Masliah E (2013) The
195. Prouteau M, Loewith R (2018) Regulation of cellular metabo- many faces of α-synuclein: from structure and toxicity to thera-
lism through phase separation of enzymes. Biomolecules peutic target. Nat Rev Neurosci 14(1):38–48. https://doi.org/
8(4):160. https://doi.org/10.3390/biom8040160 10.1038/nrn3406
196. Shen QJ, Kassim H, Huang Y, Li H, Zhang J, Li G, Wang PY, 209. Wu KP, Weinstock DS, Narayanan C, Levy RM, Baum J (2009)
Yan J, Ye F, Liu JL (2016) Filamentation of metabolic enzymes Structural reorganization of α-Synuclein at low pH observed
in Saccharomyces cerevisiae. J Genet Genomics 43(6):393– by NMR and REMD simulations. J Mol Biol 391(4):784–796.
404. https://doi.org/10.1016/j.jgg.2016.03.008 https://doi.org/10.1016/j.jmb.2009.06.063
197. Narayanaswamy R, Levy M, Tsechansky M, Stovall GM, 210. Ballatore C, Lee VMY, Trojanowski JQ (2007) Tau-mediated
O’Connell JD, Mirrielees J, Ellington AD, Marcotte EM neurodegeneration in Alzheimer’s disease and related disor-
(2009) Widespread reorganization of metabolic enzymes into ders. Nat Rev Neurosci 8(9):663–672. https://doi.org/10.1038/
reversible assemblies upon nutrient starvation. Proc Natl Acad nrn2194
Sci USA 106(25):10147–10152. https://doi.org/10.1073/pnas. 211. Williams DR (2006) Tauopathies: classification and clinical
0812771106 update on neurodegenerative diseases associated with micro-
198. Stansfield I, Jones KM, Kushnirov VV, Dagkesamanskaya tubule-associated protein tau. Intern Med J 36(10):652–660.
AR, Poznyakovski AI, Paushkin SV, Nierras CR, Cox BS, Ter- https://doi.org/10.1111/j.1445-5994.2006.01153.x
Avanesyan MD, Tuite MF (1995) The products of the SUP45 212. Drubin DG, Kirschner MW (1986) Tau protein function in
(eRF1) and SUP35 genes interact to mediate translation termina- living cells. J Cell Biol 103(6):2739–2746. https://doi.org/10.
tion in Saccharomyces cerevisiae. EMBO J 14(17):4365–4373. 1083/jcb.103.6.2739
https://doi.org/10.1002/j.1460-2075.1995.tb00111.x 213. Patel A, Lee HO, Jawerth L, Maharana S, Jahnel M, Hein MY,
199. Zhao YG, Zhang H (2020) Phase separation in membrane biol- Stoynov S, Mahamid J, Saha S, Franzmann TM, Pozniakovski
ogy: the interplay between membrane-bound organelles and A, Poser I, Maghelli N, Royer LA, Weigert M, Myers EW,
membraneless condensates. Dev Cell 55(1):30–44. https://doi. Grill S, Drechsel D, Hyman AA, Alberti S (2015) A liquid-to-
org/10.1016/j.devcel.2020.06.033 solid phase transition of the ALS protein FUS accelerated by
200. Fujioka Y, Alam JM, Noshiro D, Mouri K, Ando T, Okada Y, disease mutation. Cell 162(5):1066–1077. https://doi.org/10.
May AI, Knorr RL, Suzuki K, Ohsumi Y, Noda NN (2020) Phase 1016/j.cell.2015.07.047
separation organizes the site of autophagosome formation. Nature 214. Burke KA, Janke AM, Rhine CL, Fawzi NL (2015) Residue-
578(7794):301–305. https://d oi.o rg/1 0.1 038/s 41586-0 20-1 977-6 by-residue view of in vitro FUS granules that bind the C-ter-
201. Yamamoto H, Fujioka Y, Suzuki SW, Noshiro D, Suzuki H, minal domain of RNA polymerase II. Mol Cell 60(2):231–241.
Kondo-Kakuta C, Kimura Y, Hirano H, Ando T, Noda NN, https://doi.org/10.1016/j.molcel.2015.09.006
Ohsumi Y (2016) The intrinsically disordered protein Atg13 215. Iwabuchi K, Li B, Massa HF, Trask BJ, Date T, Fields S
mediates supramolecular assembly of autophagy initiation com- (1998) Stimulation of p53-mediated transcriptional activation
plexes. Dev Cell 38(1):86–99. https://doi.org/10.1016/j.devcel. by the p53-binding proteins, 53BP1 and 53BP2. J Biol Chem
2016.06.015 273(40):26061–26068. https://d oi.o rg/1 0.1 074/j bc.2 73.4 0.
202. Fujioka Y, Suzuki SW, Yamamoto H, Kondo-Kakuta C, Kimura 26061
Y, Hirano H, Akada R, Inagaki F, Ohsumi Y, Noda NN (2014) 216. Panier S, Boulton SJ (2014) Double-strand break repair: 53BP1
Structural basis of starvation-induced assembly of the autophagy comes into focus. Nat Rev Mol Cell Biol 15(1):7–18. https://
initiation complex. Nat Struct Mol Biol 21(6):513–521. https:// doi.org/10.1038/nrm3719
doi.org/10.1038/nsmb.2822 217. Mackay JA, Callahan DJ, Fitzgerald KN, Chilkoti A (2010)
203. Guillén-Boixet J, Kopach A, Holehouse AS, Wittmann S, Jah- Quantitative model of the phase behavior of recombinant
nel M, Schlüßler R, Kim K, Trussina IREA, Wang J, Mateju D, pH-responsive elastin-like polypeptides. Biomacromol
Poser I, Maharana S, Ruer-Gruß M, Richter D, Zhang X, Chang 11(11):2873–2879. https://doi.org/10.1021/bm100571j
YT, Guck J, Honigmann A, Mahamid J, Hyman AA, Pappu RV,
13
218. Liu W, MacKay JA, Dreher MR, Chen M, McDaniel JR, Sim- 229. Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim
nick AJ, Callahan DJ, Zalutsky MR, Chilkoti A (2010) Inject- HJ, Mittag T, Taylor JP (2015) Phase separation by low com-
able intratumoral depot of thermally responsive polypeptide- plexity domains promotes stress granule assembly and drives
radionuclide conjugates delays tumor progression in a mouse pathological fibrillization. Cell 163(1):123–133. https://doi.org/
model. J Control Release 144(1):2–9. https://doi.org/10.1016/j. 10.1016/j.cell.2015.09.015
jconrel.2010.01.032 230. Iwabuchi K, Bartel PL, Li B, Marraccino R, Fields S (1994) Two
219. Lim DW, Nettles DL, Setton LA, Chilkoti A (2008) In situ cellular proteins that bind to wild-type but not mutant p53. Proc
cross-linking of elastin-like polypeptide block copolymers for Natl Acad Sci USA 91(13):6098–6102. https://doi.org/10.1073/
tissue repair. Biomacromol 9(1):222–230. https://doi.org/10. pnas.91.13.6098
1021/bm7007982 231. Cuella-Martin R, Oliveira C, Lockstone HE, Snellenberg S, Grol-
220. Betre H, Ong SR, Guilak F, Chilkoti A, Fermor B, Setton LA musova N, Chapman JR (2016) 53BP1 integrates DNA repair
(2006) Chondrocytic differentiation of human adipose-derived and p53-dependent cell fate decisions via distinct mechanisms.
adult stem cells in elastin-like polypeptide. Biomaterials Mol Cell 64(1):51–64. https://doi.org/10.1016/j.molcel.2016.08.
27(1):91–99. https://doi.org/10.1016/j.biomaterials.2005.05. 002
071 232. Bi J, Huang A, Liu T, Zhang T, Ma H (2015) Expression of
221. McHale MK, Setton LA, Chilkoti A (2005) Synthesis and in vitro DNA damage checkpoint 53BP1 is correlated with prognosis,
evaluation of enzymatically cross-linked elastin-like polypeptide cell proliferation and apoptosis in colorectal cancer. Int J Clin
gels for cartilaginous tissue repair. Tissue Eng 11(11–12):1768– Exp Pathol 8(6):6070–6082
1779. https://doi.org/10.1089/ten.2005.11.1768 233. Cao X, Jin X, Liu B (2020) The involvement of stress granules in
222. Banani SF, Lee HO, Hyman AA, Rosen MK (2017) Biomolecu- aging and aging-associated diseases. Aging Cell 19(4):e13136.
lar condensates: organizers of cellular biochemistry. Nat Rev Mol https://doi.org/10.1111/acel.13136
Cell Biol 18(5):285–298. https://doi.org/10.1038/nrm.2017.7 234. Franzmann TM, Alberti S (2019) Prion-like low-complexity
223. Shin Y, Brangwynne CP (2017) Liquid phase condensation in sequences: key regulators of protein solubility and phase behav-
cell physiology and disease. Science 357(6357):eaaf4382. https:// ior. J Biol Chem 294(18):7128–7136. https://doi.org/10.1074/
doi.org/10.1126/science.aaf4382 jbc.TM118.001190
224. Memisoglu G, Eapen VV, Yang Y, Klionsky DJ, Haber JE (2019) 235. Kroschwald S, Maharana S, Mateju D, Malinovska L, Nuske E,
PP2C phosphatases promote autophagy by dephosphorylation of Poser I, Richter D, Alberti S (2015) Promiscuous interactions
the Atg1 complex. Proc Natl Acad Sci U S A 116(5):1613–1620. and protein disaggregases determine the material state of stress-
https://doi.org/10.1073/pnas.1817078116 inducible RNP granules. Elife 4:e06807. https://d oi.o rg/1 0.7 554/
225. Takahara T, Maeda T (2012) Transient sequestration of TORC1 eLife.06807
into stress granules during heat stress. Mol Cell 47(2):242–252. 236. Lobaskin V, Qamhieh K (2003) Effective macroion charge and
https://doi.org/10.1016/j.molcel.2012.05.019 stability of highly asymmetric electrolytes at various salt con-
226. Yang YS, Kato M, Wu X, Litsios A, Sutter BM, Wang Y, Hsu ditions. J Phys Chem B 107(32):8022–8029. https://doi.org/10.
CH, Wood NE, Lemoff A, Mirzaei H, Heinemann M, Tu BP 1021/jp027608+
(2019) Yeast Ataxin-2 Forms an Intracellular Condensate 237. Zhang FJ, Roosen-Runge F, Sauter A, Wolf M, Jacobs RMJ,
Required for the Inhibition of TORC1 Signaling during Respira- Schreiber F (2014) Reentrant condensation, liquid-liquid phase
tory Growth. Cell 177(3):697-710.e17. https://d oi.o rg/1 0.1 016/j. separation and crystallization in protein solutions induced by
cell.2019.02.043 multivalent metal ions. Pure Appl Chem 86(2):191–202. https://
227. Wippich F, Bodenmiller B, Trajkovska MG, Wanka S, Aebersold doi.org/10.1515/pac-2014-5002
R, Pelkmans L (2013) Dual specificity kinase DYRK3 couples
stress granule condensation/dissolution to mTORC1 signaling. Publisher's Note Springer Nature remains neutral with regard to
Cell 152(4):791–805. https://doi.org/10.1016/j.cell.2013.01.033 jurisdictional claims in published maps and institutional affiliations.
228. Wheeler JR, Matheny T, Jain S, Abrisch R, Parker R (2016)
Distinct stages in stress granule assembly and disassembly. Elife
5:e18413. https://doi.org/10.7554/eLife.18413
13
1. use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
2. use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
3. falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
4. use bots or other automated methods to access the content or redirect messages
5. override any security feature or exclusionary protocol; or
6. share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com