Ladecola C, Anrather J. (2012)
Ladecola C, Anrather J. (2012)
Ladecola C, Anrather J. (2012)
Immunity and inflammation are key elements of the pathobiology of stroke, a devastating illness second only to cardiac ischemia
as a cause of death worldwide. The immune system participates in the brain damage produced by ischemia, and the damaged
brain, in turn, exerts an immunosuppressive effect that promotes fatal infections that threaten the survival of people after stroke.
© 2011 Nature America, Inc. All rights reserved.
Inflammatory signaling is involved in all stages of the ischemic cascade, from the early damaging events triggered by arterial
occlusion to the late regenerative processes underlying post-ischemic tissue repair. Recent developments have revealed that
stroke engages both innate and adaptive immunity. But adaptive immunity triggered by newly exposed brain antigens does not
have an impact on the acute phase of the damage. Nevertheless, modulation of adaptive immunity exerts a remarkable protective
effect on the ischemic brain and offers the prospect of new stroke therapies. As immunomodulation is not devoid of deleterious
side effects, a better understanding of the reciprocal interaction between the immune system and the ischemic brain is essential
to harness the full therapeutic potential of the immunology of stroke.
Inflammation affects the brain after stroke, and cells of the immune cytotoxic response against newly exposed brain antigens and their role
system, such as neutrophils and macrophages, have traditionally been in the acute and chronic phase of the injury. Finally, we will evaluate the
used by neuropathologists and forensic pathologists to determine the therapeutic opportunities afforded by modulation of the immune system
approximate age of cerebrovascular lesions1. Although inflammation and their potential pitfalls.
was commonly thought to be merely a reaction to tissue damage, it has
been increasingly recognized as a key contributor to the pathophysi- Inflammatory signaling in the early post-ischemic period
ology of cerebrovascular diseases, especially stroke caused by arterial Post-ischemic inflammation is characterized by an orderly sequence of
occlusion or ischemic stroke2. Recent evidence suggests that elements events involving the brain, its vessels, the circulating blood and lymphoid
of the immune system are intimately involved in all stages of ischemic organs. Inflammation is an integral part of the cascade of events triggered
cascade (Box 1), from the acute intravascular events triggered by the by ischemia and reperfusion (Box 1). The inflammatory process begins
interruption of the blood supply to the parenchymal processes leading in the intravascular compartment immediately after arterial occlusion,
to brain damage and the ensuing tissue repair. In turn, the ischemic when the ensuing hypoxia, changes in shear stress and production of
brain, through the autonomic nervous system, exerts a potent suppres- reactive oxygen species (ROS) trigger the coagulation cascade, and lead
sive effect on lymphoid organs that promotes intercurrent infections, to activation of complement, platelet and endothelial cells6–9(Fig. 1).
a major determinant of stroke morbidity and mortality3,4. Therefore, The resulting intravascular formation of fibrin traps platelets and leu-
the immune system is closely involved on determining the fate of the kocytes, leading to microvascular occlusions10,11. Within minutes after
ischemic brain and the survival of people after stroke. In multiple scle- ischemia, the adhesion molecule P-selectin is translocated to the surface
rosis, the classical inflammatory disease of the central nervous system membrane of platelets and endothelial cells12, and proinflammatory sig-
(CNS), elements of innate and adaptive immunity are engaged in the nals are rapidly generated (Table 1). Oxidative stress in endothelial cells
post-ischemic brain5. Thus, molecular cues generated by cerebral isch- reduces the bioavailability of nitric oxide (NO), which is a potent vaso-
emia activate components of innate immunity, promote inflammatory dilator and an inhibitor of platelet aggregation and leukocyte adhesion12.
signaling and contribute to tissue damage. At the same time, these Loss of the beneficial effects of NO exacerbates intravascular plugging
processes stimulate a potentially damaging adaptive immune response and aggravates the ischemic insult by reducing blood flow to the ische-
directed at antigens previously sequestered behind the blood-brain bar- mic territory13,14. Furthermore, oxidative stress leads to constriction of
rier (BBB). These recent developments warrant a reevaluation of the pericytes, contractile cells that replace myocytes in capillaries, producing
contribution of inflammation and immunity to stroke pathophysiol- more microvascular occlusions15. Oxidative stress and inflammatory
ogy. This review will discuss the involvement of innate and adaptive mediators also alter the permeability of the BBB, which increases the
immunity in ischemic brain injury and their impact on tissue damage number of pinocytotic vesicles in the cytoplasm of endothelial cells,
and repair. Furthermore, we will examine the evidence for an adaptive enhancing transendothelial transport16. Proteases are expressed in vas-
cular cells and released by leukocytes, whereas junctional proteins that
Division of Neurobiology, Department of Neurology and Neuroscience, Weill seal adjacent endothelial cells are downregulated, facilitating extravasa-
Cornell Medical College, New York, New York, USA. tion of proteins and cells through the paracellular route (Fig. 1)16. In
Correspondence should be addressed to C.I. (coi2001@med.cornell.edu). the perivascular space, ischemia and reperfusion activate perivascular
Platelet
Intravascular
P-selectin activation C3a
Ischemia-- PSGL-1 Mac-1
reperfusion
n Leukocyte
y AA Protease
Prote ase
Hypoxia, ROS,
OS, release
relea
ase
shear stresss IL-1a LFA-1
Vascular wall
EC Tight junction IICAM-1 NO
Open BBB ROS
RO
O
Myocyte
Perivascular
P2X7 Histamine
space
proteases
TNF IL-1b TNF CD8888
C5a
Macrophage Mast cell Glu Astrocyte
Brain parenchyma
CX3CL1
Glu
CD200 CX3CR1
GluR1
Neuron CD200R ATP
Katie Vicari
P2X7
P2Y2 TNF
ATP IL-1b
Microglia ROS
© 2011 Nature America, Inc. All rights reserved.
Figure 1 Early vascular, perivascular and parenchymal events triggered by ischemia and reperfusion. Hypoxia, ROS and changes in shear stress initiate the
cellular events induced by ischemia and reperfusion. In the vessels lumen, ischemia and reperfusion lead to blood clotting, platelet aggregation and cytokine
(IL-1a) release. Translocation of P-selectin on the surface of platelets and endothelial cells leads to platelet-leukocyte aggregation. Complement is activated,
and arachidonic acid metabolites (AA) are released. In the vascular wall, upregulation of E- and P-selectin on endothelial cells provides a platform for low
affinity leukocyte binding through interaction with glycoproteins expressed on leukocytes, for example, P-selectin glycoprotein ligand-1. Firm adhesion
is obtained after endothelial expression of ICAM-1 interacting with leukocyte b2 integrins (LFA-1 and Mac-1). Loss of NO promotes vasoconstriction and
enhances leukocyte and platelet aggregation. MMP activation could lead to BBB breakdown and matrix proteolysis, facilitating leukocyte extravasation. In
the perivascular space, chemotactic complement subunits (C5a) acting on mast cell complement receptors (CD88) leads to degranulation and release of
histamine and proteases, contributing to BBB leakiness. Cytokines (TNF, IL-1b) are produced by mast cells and perivascular macrophages, providing further
signals to guide leukocyte migration across the vessel wall. In the brain parenchyma, injured cells release purines (ATP), which act as early proinflammatory
signals leading to production of cytokines and chemokines. Disruption of neuronal-microglial interaction (CX3CL1, CD200) and increases in extracellular
glutamate (Glu) acting on microglial GluR1 metabotropic receptor27 also contribute to the proinflammatory milieu.
macrophages and mast cells. Mast cell degranulation releases vasoac- (Fig. 1), activation of these receptors downregulates microglial cytokine,
tive mediators, such as histamine, proteases and tumor necrosis factor ROS and NO production27 and suppresses the secretory response in
(TNF), whereas activated macrophages release proinflammatory cyto- mast cells28,29. Therefore, ATP represents an early neuronal danger sig-
kines17–20 (Box 2) (Fig. 1). These proinflammatory mediators contribute nal, promoting the inflammatory response of resident immune cells,
to the endothelial expression of adhesion molecules and to the BBB whereas neurotransmitter release may oppose these changes and coun-
damage that promotes the infiltration of leukocytes (neutrophils, lym- teract inflammation.
phocytes and monocytes)19. Cell death and pattern recognition receptors in the post-ischemic
brain. A different signaling landscape emerges after cells begin to die.
Ischemic cell death sets the stage for innate and adaptive A wide variety of molecular signals is released from the intracellular
immunity compartment or is generated by the action of lytic enzymes escaped
As the ischemic cascade progresses (Box 1), cell death leads to a new from dead cells on matrix proteins (Fig. 2). These so-called danger-
phase of the inflammatory response (Fig. 2). Dying and dead cells release associated molecular pattern molecules (DAMPs) activate pattern
‘danger signals’ that activate the immune system21. Some of these signals, recognition receptors, including Toll-like receptors (TLRs) and scav-
such as the nucleotides ATP and UTP, are released by cells under stress enger receptors, which are widely expressed on microglia, perivascular
when the cell membrane is still intact and set the stage for the subsequent macrophages and brain endothelial cells30. DAMPs and purines act
immune response22. in concert to induce the expression of proinflammatory molecules in
ATP and neurotransmitters. Extracellular ATP abundance increases infiltrating leukocytes (Table 1) and to prime dendritic cells (DCs) for
within minutes after ischemia as a result of neuronal and glial depolar- antigen presentation (Boxes 2 and 3) (Fig. 2). Considering the high
ization or escape through damaged plasma membranes23–25. ATP is also vascular density of the brain, inflammatory mediators released from
released by vascular cells and blood cells and may promote intravascular parenchymal cells are likely to feed back on the vascular and perivas-
coagulation and platelet aggregation26. High parenchymal ATP amounts cular compartments to reinforce and amplify the expression of cytok-
activate P2X7 receptors in microglia, leading to release of proinflam- ines, chemokines and adhesion molecules that drive the infiltration of
matory mediators (Fig. 2). Activated microglia develop many charac- blood-borne cells into the ischemic tissue. After neuronal death, loss of
teristics of macrophages, including ameboid morphology, migratory cell-to-cell interaction between neurons and microglia, which are kept
capacity, phagocytosis and major histocompatibility complex (MHC) quiescent in the normal state by contact with neurons, also promotes
class II–restricted antigen presentation (Box 3). At the same time, neuro- inflammatory signaling (Fig. 1). For example, CD200, a surface protein
transmitters release after ischemia and reperfusion may counteract the expressed in neurons, interacts with its receptor CD200R on microglia,
emerging inflammatory response. Microglia express a wide variety enforcing a resting phenotype31. Disruption of this interaction due to
of neurotransmitter receptors, including AMPA, kainate, adrenergic, post-ischemic loss of CD200 (ref. 32) may promote microglial activa-
GABAB opioid and cannabinoid receptors27. With some exceptions27 tion (Fig. 1). Similarly, CX3CL1 (fractalkine), a cell surface–bound
chemokine constitutively expressed by neurons, suppresses microglial tion5. Growing evidence also indicates adaptive immunity in stroke.
activation through its microglial receptor CX3CR1. Thus, after neu- Stroke and adaptive immunity. Antibodies against CNS antigens
ronal injury, loss of CX3CL1 results in enhanced microglial activation develop after ischemic stroke, suggesting a humoral immune response
in several inflammatory disease models33. In addition, increasing con- to the injury, and circulating T cells become sensitized against CNS
centrations of extracellular glutamate activate metabotropic glutamate antigens, such as myelin basic protein (MBP) and related peptides
receptors on microglia, leading to a proinflammatory phenotype34 (Table 2). Antigen-presenting cell (APC) numbers are reduced in the
(Fig. 1). Therefore, as neuronal death develops in the ischemic core periphery and increased in the ischemic brain both in rodent and human
and spreads to the penumbra, the loss of the immunosuppressive effect stroke35–38. The accumulation of APCs coincides with the peak of lym-
afforded by neurotransmitter release and neuron-microglia interaction phocytic infiltration and is associated with expression of MHC class II
may also foster post-ischemic inflammation. molecules and the co-stimulatory molecule CD8035,37,38, findings sug-
Collectively, these observations suggest that the inflammatory gestive of antigen presentation (Box 3). Support for involvement of adap-
response after ischemia and reperfusion starts at the vascular level, tive immunity also comes from studies on the role of lymphocytes in
driven by nontranscriptional events triggered by hypoxia, shear stress rodent models of focal cerebral ischemia. Ischemia leads to infiltration
and ROS production. As ischemia damages the brain tissue, danger of the major lymphocytes subtypes into the ischemic brain37 (Box 3),
signals are released first from cells under stress and then from necrotic and increasing evidence suggests that they contribute to ischemic
cells. Concomitant with the loss of immunosuppressive mechanisms, injury (Table 2 and Fig. 3). In fact, lymphocyte-deficient mice are pro-
these signals activate purinergic receptors and pattern recognition tected from ischemic damage39,40. The protection has been attributed
receptors, which induce an inflammatory response in resident brain to T cells, because B cell–deficient mice or lymphocyte-deficient mice
cells and infiltrating leukocytes, resulting in inflammation-induced cell reconstituted with B cells are still protected from injury (Table 2). gdT
death (Box 4). cells also have been shown to contribute to the injury by releasing the
proinflammatory cytokine IL-17 (ref. 41) (Fig. 3). In contrast, T regu-
Does adaptive immunity contribute to ischemic brain injury? latory (Treg) cells are protective in the late stage of cerebral ischemia,
Danger signals released from damaged cells also promote the presen- an effect evident only if the injury is small42,43. Additional evidence in
tation of tissue antigens that were previously hidden by the BBB or favor of the involvement of cell-based adaptive mechanisms in stroke
that develop as a result of the breakdown of cell membranes21 (Fig. 2). damage was provided by studies in which animals were tolerized against
Antigen presentation leads to the development of cellular and humoral myelin-derived peptides (Table 2). Repeated mucosal administration of
immunity directed against the antigens (Box 3). This adaptive immune myelin antigens (tolerization) before arterial occlusion protects rodents
response has the potential of inducing autoimmunity against the organ from ischemic brain injury44. Although tolerization is antigen specific,
in which the cell death occurred, as described in the heart (Dressler’s its beneficial effects are not restricted to immune responses directed
syndrome), eye (sympathetic ophthalmia) and pancreas (diabetes)21. at the inducing antigen, but are instead more widespread, a phenom-
Furthermore, the damaging effect of adaptive immunity is well estab- enon termed bystander suppression44. The protection can be induced
lished in multiple sclerosis and in models of autoimmune demyelina- in naive mice by adoptive transfer of splenocytes or CD4+ T cells from
ROS on lipids. The cytokine production and complement activation resulting from these events leads to increased leukocyte infiltration and enhances tissue
damage, which, in turn, produces more DAMPs. Antigens unveiled by tissue damage are presented to T cells, setting the stage for adaptive immunity.
tolerized animals45,46, suggesting the involvement of cellular immune result from suppression of autoreactive T cells targeted against myelin49.
mechanisms. Examination of T cell function indicated that activation Although RTLs may not bind T cells—but rather APC and platelets50—
of tolerized T cells by the antigen unveiled by the stroke induces a TH2 the findings suggest a role of cellular immunity in the mechanisms of
cytokine response (Box 3)45,47,48. The effect has been attributed to IL-4 ischemic brain injury.
and IL-10 production, which favors the formation of transforming Despite the evidence supporting an autoimmune response against
growth factor-b (TGF-b)-secreting Treg cells45,46,48. Other studies have the post-ischemic brain, there are inconsistencies with the hypothesis
found that administration of recombinant T cell receptor ligand (RTL), that classical adaptive immunity contributes ischemic brain injury. The
consisting of a1 and b1 domains of MHC class II complex bound to a temporal profile of the involvement T cells in brain damage is not con-
myelin peptide antigen (myelin oligodendrocyte glycoprotein (MOG)- sistent with established concepts of adaptive immunity51 (Fig. 3). Thus,
35–55), reduces stroke volume in focal ischemia49. The protective effect the protective effect observed in lymphocyte-deficient mice or afforded
is associated with reduction of infiltrating inflammatory cells and may by blocking postischemic trafficking of T cells into the ischemic brain
Table 2 Selected evidence for and against the involvement of adaptive immunity in ischemic brain injury
In favor of adaptive immunity causing tissue damage
Evidence Findings References
CNS antigens and associated humoral response are present after MBP, NSE, S100b, GFAP, NMDA receptor, neurofilament 117–119
stroke
T cell response to CNS antigens after stroke Lymphocyte sensitization to CNS antigens 120
APC increase in the human and rodent brain after stroke DC and macrophages found in perivascular space and brain parenchyma after stroke 35–38
gdT cells and Treg cells are involved in experimental stroke gdT cells contribute to brain damage through IL-17; Treg cells are protective in the 41,42
late phase of cerebral ischemia
T cells sensitized against CNS antigens mediate damage in stroke RTL targeted to myelin-specific T cells reduces ischemic brain injury 49
Tolerization to CNS antigens is protective in experimental stroke Mucosal administration of MBP or MOG reduces damage in focal ischemia 44
Against adaptive immunity causing damage
Evidence Findings References
Temporal dissociation between adaptive response and tissue T cell–mediated damage occurs early (<24 h) after stroke, not consistent with anti- 39,52
damage gen presentation and clonal expansion
T cell–mediated ischemic damage is antigen independent T cells reactive against CNS or non-CNS antigens are equally damaging 40
Absence of co-stimulatory molecules necessary for antigen pre- Mice lacking CD28 or B7 are not protected from focal ischemia 40
sentation does not affect stroke outcome
Unlike other models of autoimmunity, both CD4+ and CD8+ T CD4- and CD8-null mice are equally protected 41,52
cells are involved in the injury
© 2011 Nature America, Inc. All rights reserved.
CD, cluster of differentiation; GFAP, glial fibrillary acidic protein; MBP, myelin basic protein; NSE, neuron specific enolase.
occurs 24–48 h after ischemia40, whereas adaptive responses require an thrombotic actions leading to microvascular occlusions52. These effects
interval of 7–10 d from antigen presentation to the clonal expansion of would be deleterious by preventing reperfusion and by compromising
autoreactive T cells and immune attack on the target organ51 (Box 3). collateral flow after ischemia and reperfusion. But lack of lymphocytes
Furthermore, reconstitution of lymphocyte-deficient mice with T cells does not improve post-ischemic cerebral blood flow at least in the acute
targeting non-CNS antigens worsens ischemic damage, and mice lacking phase53, nor does it suppress thrombus formation40. Therefore, effects
co-stimulatory molecules essential for antigen-specific T cell response of lymphocytes altering microvascular perfusion or patency seem
are not protected from ischemia40. It is also surprising that, unlike auto- unlikely. Another possibility is that natural killer T (NKT) or gdT cells,
immune responses in other organs where there is a prevalence for either T cells that have a simplified T cell receptor (TCR) and may not require
T helper or T effector cell participation, both CD4+ and CD8+ T cells antigen processing and MHC presentation, as well as NK cells (Box 3),
are involved in ischemic injury52. Collectively, these observations argue are responsible for these early cytotoxic effects of lymphocytes. In sup-
against an autoimmune attack on the brain resulting from presentation port of this hypothesis, NKT cells are not present in recombination-
of CNS antigen released by the stroke. activating gene 1 (Rag1)-/- mice and severe combined immunodeficient
The lymphocyte puzzle. If lymphocytes do not mediate an autoim- mice54, and they could contribute to explain the early neuroprotection
mune attack on the brain, how do they contribute to the early phase of observed in these models of lymphocyte deficiency39,40. However, CD1-
ischemic brain injury? One possibility is that these cells participate in deficient mice lack NKT cells and are not protected from ischemic
the cerebrovascular dysfunction occurring after ischemia or have pro- injury 24 h after middle cerebral artery occlusion40, suggesting that
NKT cells may not be involved in the early phase of the injury. gdT
Acute phase of ischemia Delayed phase of ischemia cells have been implicated in ischemic brain injury, but their involve-
ment seems restricted to the late phase of cerebral ischemia (4 d)40,41.
Unprimed γδ CD4+ Helper CD8+
function Considering the limited number of studies available, the involvement
T cell T cell T cell T cell
of NK, NKT and gdT cells, lymphocyte subtypes that act in a fashion
ROS?
IFN-γ? IL-17 akin to innate immunity, needs further exploration.
Detrimental
CNS antigens
The resulting cell death could play a part in the delayed brain damage and
Innate Adaptive atrophy that occur after stroke83.
their processes. Microglia contribute to post-ischemic inflammation and secretory vesicles. Major proinflammatory molecules
by producing TNF, IL-1b, ROS and other proinflammatory mediators stored in granules and vesicles include iNOS, NADPH oxidase,
(Table 1 and Fig. 1). However, they also contribute to the resolution myeloperoxidase, MMP-8, MMP-9, elastase and cathepsins.
of inflammation and tissue repair by producing IL-10 and TGF-b, as After cerebral ischemia, neutrophils adhere to the cerebral
well as several growth factors, including IGF-1 (Table 1 and Fig. 4). endothelium and transmigrate into the tissue12. Vesicle and
granule exocytosis is induced after receptor engagement,
Perivascular macrophages are confined to the space between such as binding to E-selectin on endothelial cells and IL-8
the vascular basement membrane and the brain surface (glia stimulation129.
limitans) and, unlike microglia, are continuously replenished by
hematogenous precursors126. Macrophages can be classified Is the immune system comparable in rodents and humans?
into two groups: M1 macrophages that produce proinflammatory Circulating neutrophils predominate in humans (50–70%),
cytokines (IL-1b, IL-12, IL-23 and TNF), chemokines, ROS whereas lymphocytes predominate in rodents (75–90%) 130.
and NO, promoting a TH1 immune response (Box 3), and M2 Several immune-related molecules, such as iNOS, P2X7,
macrophages that produce anti-inflammatory cytokines (IL-10 and TLR2, antigen-presenting proteins, co-stimulatory molecules,
TGF-b), IL-1ra and arginase127. After cerebral ischemia, cytokines immunoglobulins and chemokines, are differentially expressed in
produced by perivascular macrophages are thought to drive the mice and humans or present in one species and not the other 130.
infiltration of inflammatory cells19. Human endothelial cells can present antigens to resting CD4 +
and CD8+ T cells, whereas mouse endothelial cells can only
Mast cells are found in meninges and cerebral blood vessels. Mast activate CD8+ T cells131. Consequently, the immune responses
cells granules store vasoactive substances (histamine), cytokines initiated by vascular pathologies may differ quantitatively and
(TNF), anticoagulants (heparin) and proteases (tryptase, chymase, qualitatively between humans and mice. The impact of these
MMP2 and MMP9)17,18. In addition, mast cells are capable of differences needs to be considered when translating findings
phagocytosis and antigen presentation and can modulate the obtained from rodent models of cerebral ischemia to human
adaptive immune response128. stroke.
Collectively, these studies suggest that, although an antigen-specific reorganization of the injured brain. The factors governing resolution
immune response may develop after stroke, evidence showing that of inflammation and the reestablishment of tissue homeostasis are still
autoreactive T cells can attack the brain tissue after CNS antigens poorly understood, particularly in brain. Increasing evidence suggests
against which they were sensitized are exposed by ischemic damage is that resolution of inflammation is not a passive process due to exhaus-
lacking. Lymphocytes have a role in the development and progression tion of the signaling; instead, it is orchestrated by the interplay of a large
of the injury, but how they exert their powerful effect does not conform number of mediators that actively suppress the inflammatory response55.
to the tenets of classical autoimmunity. The role of NK, NKT and gdT Major steps in the process include removal of dead cells, development
cells, which could contribute to the acute phase of the injury, needs of an anti-inflammatory milieu and generation of prosurvival factors
further exploration. Similarly, considering the evidence for humoral fostering tissue reconstruction and repair55,56.
immune responses in stroke (Table 2), the contribution of B cells to Clearing dead cells. Microglia and infiltrating macrophages consti-
the damage needs a more in-depth assessment. tute the predominant phagocytes removing dead cells and tissue debris
after stroke57,58, a process orchestrated by ‘find me’ and ‘eat me’ sig-
Resolution of inflammation and tissue repair nals. Find-me signals, including purines released from injured cells and
Post-ischemic inflammation is a self-limiting process that eventually chemokines, attract microglia and macrophages to the site of injury59,60
subsides and prepares the terrain for the structural and functional (Fig. 4). These phagocytic cells are then presented with eat-me signals
Post-ischemic
Clearing dead cells inflammation resolution Brain repair
VEGF
Antigen
presentation Neuron
‘Find me’
Dying UTP, ATP, MMPs
neuron P2Y2
chemokines
DC
PtdSer Blood
BDNF BDNF vessel
Neutrophil G-CSF VEGF
Katie Vicari
cytokines
Neuroprotective IL-1, TNF
© 2011 Nature America, Inc. All rights reserved.
Figure 4 Resolution of inflammation and tissue repair. Clearing of dead cells and suppression of inflammation are key events in brain repair. Find-me signals
(UTP, ATP) attract microglia and macrophages through P2Y2 receptors. Eat-me signals include UDP, which acts on P2Y6 receptors to stimulate microglial
phagocytosis112, and phosphatidylserine (PtdSer), which is translocated to the outer leaflet of the plasma membrane of apoptotic cells 112. PtdSer-binding
proteins involved in the clearance of dead cells include milk fat globule epidermal growth factor 8 protein on microglia 113 and T cell immunoglobulin and
mucin domain-containing molecule 4 (TIM4) on macrophages112. Immunoglobulins directed against CNS antigens, which appear after stroke (Table 2), may
also promote phagocytosis by engaging Fc receptors on phagocytic cells. Phagocytosis promotes secretion of IL-10 and TGF-b 56, which, in turn, suppress
antigen presentation, promote Treg formation, inhibit expression of adhesion molecules in endothelial cells and production of proinflammatory cytokines 61.
TGF-b and IL-10 are also neuroprotective114,115 and may facilitate brain repair processes. In addition, lipoxins, resolvins and protectins, metabolites of
arachidonic acid and omega-3 fatty acids that play an active part in the resolution of inflammation in other organs 55, could also contribute to suppress post-
ischemic inflammation. Growth factors and MMPs produced by endothelial cells, neurons, astrocytes, oligodendrocytes and microglia are key molecules
driving tissue reorganization and repair63,116.
associated with dying or dead cells (Fig. 4). recovery has also been highlighted by studies in which the transcriptome
TGF-β, IL-10 and the anti-inflammatory milieu. TGF-b and IL-10 of sprouting neurons indicated involvement of MHC I class molecules
are pleiotropic immunoregulatory cytokines that have a crucial role in and complement subunits66.
the development of the anti-inflammatory milieu associated with tis- This evidence indicates that cells of the immune system have a fun-
sue repair (Fig. 4). The production of these cytokines is promoted by damental role in all the phases of post-ischemic brain recovery. But the
phagocytosis and occurs in concert with the removal of dead cells56. limited data available provide only a glimpse into the complex sequence
TGF-b, which is upregulated after ischemia primarily in microglia and of events that reestablish the structural and functional homeostasis of
macrophages, has neuroprotective properties and profound effects on the brain after stroke. Additional studies on recently identified media-
immune cells. Although TGF-b is well known for its proinflammatory tors instrumental to inflammation resolution and tissue repair, such as
effects, it can also suppress inflammation by inhibiting T helper type lipoxins, resolvins, protectins, progranulins and cyclopentenone prosta-
1 (TH1) and TH2 responses and promoting Treg cell development61. glandins (Table 1 and Fig. 4)55,56, are needed to fully elucidate the role
Similarly, the immunoregulatory cytokine IL-10, produced by multiple of the immune system in brain repair after stroke.
cell types, including Treg cells42, has both neuroprotective and anti-
inflammatory activities (Fig. 4). Therefore, post-ischemic production of Stroke and systemic immunity
TGF-b and IL-10 can facilitate tissue repair by promoting the resolution Concomitant with the inflammatory response involving the brain,
of inflammation and exerting direct cytoprotective effects on surviving immunological changes are also observed in the blood, bone marrow,
cells in the ischemic territory. spleen and other lymphoid organs3,73. Genome profiling of peripheral
Growth factors. Post-ischemic production of growth factors helps to blood in individuals after stroke has demonstrated characteristic pat-
establish an environment that is favorable to neuronal sprouting, neu- terns of inflammatory gene expression that can help determine the cause
rogenesis, angiogenesis, gliogenesis and matrix reorganization62–64. of ischemic stroke74, reflecting the specificity of the systemic inflam-
Inflammatory cells, as well as neurons and astrocytes65,66, are capable matory response to brain injury. In rodent models, as in people with
of producing a vast array of growth factors (Table 1). For example, ischemic stroke, white blood cell count and expression of cytokines and
microglia are required for the full expression of insulin-like growth inflammatory markers are increased within hours after ischemia75–78.
factor 1 (IGF-1)67, a key factor in post-ischemic neuronal sprouting66, Such an acute-phase response is followed within 1 or 2 d by a marked
whereas reactive astrocytes are required for functional recovery after immunodepression, especially in individuals with large strokes, char-
stroke68. Vascular endothelial growth factor (VEGF), which is crucial acterized by lymphopenia, reduced functional activity of monocytes,
for post-ischemic angiogenesis, is produced by reactive astrocytes69, and upregulation of anti-inflammatory cytokines, lymphocyte apoptosis and
its action may require neutrophil matrix metalloproteinases (MMPs), splenic atrophy78,79. These immunological changes are associated with
suggesting a link between inflammatory cells and angiogenesis70. But a higher tendency to respiratory and urinary tract infections, which are
VEGF administration early after ischemia or in excessive doses may responsible for considerable morbidity and mortality in people after
enhance the damage71,72. The role of inflammatory signaling in brain stroke3,4. Infections tend to occur in patients with larger stroke and
ab chains or gd chains, immunoglobulin-like proteins consisting the membrane with perforin and inducing apoptosis via
of variable and fixed regions51. After antigen presentation, CD4+ granzyme-induced caspase activation or the Fas ligand pathway
T cells undergo clonal expansion in lymphoid organs, a process (Box 4). Lymphocytic cells with cytotoxic function also include
promoted by autocrine production of IL-2 (ref. 51). NK cells and NKT cells. NK cells lack a TCR and, as such, do not
require antigen presentation for their activation and cytotoxicity,
Helper T cells. With notable exceptions, CD4+ T cells become which is triggered by interferons or cytokines133. NKT cells have
helper T cells that do not have cytotoxic function but act as a simplified TCR that recognizes glycolipids presented by cells
‘helpers’ by coordinating and modulating immune responses132. also expressing the MHC class I–like glycoprotein CD1 (ref. 133).
Helper T cells include effector and regulatory cells132. Depending NKT cells exert their cytotoxic effect by releasing large amounts
on the molecular signals present in their milieu, different of IFN-g, IL-2 and TNF.
subpopulations of effector helper T cells can develop to produce
a specific pattern of cytokines. TH1 effector cells secrete IFN-g γδT cells. gdT cells are a subset of effector lymphocyte with a
and TNF, and their development is promoted by IL-12 through TCR comprising gd chains and particularly enriched in mucous
the transcription factor t-bet132. TH1 cells stimulate innate and T membranes134. Like NKT cells, these cells recognize nonpeptide
cell–induced immune responses leading to cytotoxicity. TH2 cells antigens and react to danger signals produced by stressed cells134.
secrete IL-4, IL-5, IL-9, IL-10 and IL-13 and promote humoral gdT cells can exert different functions depending on the context in
immunity, mucosal immunity and responses directed against which they operate, ranging from cytolysis to antigen presentation,
extracellular pathogens132. IL-4 promotes TH2 cell development, immunoregulation and production of growth factors135. In cerebral
which is regulated by the transcription factor GATA-3 (ref. 132). ischemia, gdT cells have been implicated in both cytotoxicity and
TH17 cells secrete IL-17, and their development is driven by TGF-b protective immunomodulation.
with low CD4+ lymphocyte counts and elevated plasma levels of IL-10 survivors have dementia, often associated with brain atrophy83. The
and IL-6, reflecting immunodepression80. How these systemic immune bases for the cerebral atrophy are not entirely clear, but immunological
changes are mediated is not completely understood, but there is evidence mechanisms triggered by the stroke cannot be discounted. Pathological
that sympathetic activation and the attendant release of stress steroids studies have shown an inflammatory infiltrate that persists for years
and catecholamines are involved3,4. Thus, cortisol and catecholamines after the stroke. For example, mononuclear cells, perivascular cuffing
plasma levels are elevated in people after stroke most susceptible to infec- and macrophages can be found, respectively, in 42%, 44% and 75% of
tion81, and steroid antagonists and the b-adrenergic receptor antagonist brains with stroke in a large series1, and T cells and DCs are observed
propanolol counteract lymphocyte apoptosis and infection propensity in ‘old’ strokes38. Nevertheless, these focal inflammatory changes have
after stroke in rodent models82. not been linked to post-stroke immunosuppression or dementia, and
their pathogenic significance remains uncertain.
Bright and dark sides of post-stroke immunosuppression On the other hand, immunosuppression after stroke is deleterious
What is the biological importance of immunodepression after stroke? in that it increases the incidence of infections, a major determinant of
On the one hand, the lymphopenia and immunosuppression may limit poor neurological outcome, morbidity and mortality3,4. Acute infec-
the development of autoreactive T cells targeted to CNS antigens and tion could also negatively affect stroke outcome by upregulating co-
dampen a potential autoimmune attack on the brain3,4. The relative stimulatory molecules and promoting antigen presentation21. This
rapidity of the development of ischemic brain injury is not consistent possibility is supported by studies in which bacterial lipopolysaccha-
with the temporal profile of an adaptive response against the brain; how- ride (LPS) administered at the time of reperfusion to simulate infection
ever, it is conceivable that sensitization to CNS antigens may be involved after stroke, worsens the outcome of experimental stroke84 and increases
in the long-term outcome of the stroke (Fig. 3). About 30% of stroke post-ischemic brain atrophy assessed 1 month after stroke85,86. The
effect is associated with increased expression of B7.1, a co-stimulatory tory signaling, such as complement, TLR or scavenger receptors, is
molecule needed for efficient antigen presentation (Box 3), as well as T highly protective in experimental models (Supplementary Table 1).
cell sensitization against CNS antigens and a TH1 cytokine response85,86. Furthermore, minocycline, an agent with multiple neuroprotective
Therefore, immunosuppression after stroke is detrimental by increasing actions, including broad anti-inflammatory properties, has shown
the incidence of systemic infections and, possibly, by promoting antigen promise in clinical trials95 (Table 3).
presentation and autoimmunity against the brain, which may worsen Immunomodulation and T cell–based approaches. Another strat-
the long-term sequelae of the stroke. egy is to develop approaches in which the immune system is directed
to suppress the deleterious effects of inflammation while enhancing its
Bench to bedside: trials, tribulations and promising therapies protective potential.
Ischemic stroke remains an enormous therapeutic challenge. Currently, Ischemic tolerance provides an example of protective immunomodu-
thrombolysis with tissue plasminogen activator is the only effective lation. Ischemic tolerance or preconditioning is a phenomenon in which
therapy, but, owing to the narrow therapeutic window of less than 4.5 h a sublethal injurious stimulus protects an organ against a subsequent
and safety concerns, fewer than 5% of stroke patients receive this treat- lethal stimulus. For example, a short nondamaging ischemic insult to
ment87. Among the potential therapeutic approaches targeting the the brain (local preconditioning) or other organs (remote precondition-
ischemic cascade (Box 1), preclinical studies in rodent models suggest ing) protects the brain from a subsequent damaging ischemic insult2,96.
that suppression of inflammation offers unique advantages. First, these Although ischemic tolerance is known to protect the brain by simultane-
treatments have an extended therapeutic window and are effective when ously suppressing multiple pathways in the ischemic cascade2, modula-
administered up to 12–24 h after stroke88,89. Therefore, they could be tion of the post-ischemic immune response has emerged as one of its key
used in patients who fail the time window for thrombolysis. Second, effector mechanisms30,97. Similarly, administration of low doses of LPS
as suppression of inflammation is also beneficial in models of cerebral induces tolerance and protects the brain from ischemic damage26. In this
© 2011 Nature America, Inc. All rights reserved.
hemorrhage90, concerns about worsening brain injury in patients in case, post-ischemic TLR4 signaling is redirected toward production of
whom hemorrhagic stroke has not been excluded would be minimized interferon-b (IFN-b), which, in turn, reprograms the immune system to
and early treatment by emergency medical teams would become feasible. suppress the production of proinflammatory cytokines and the infiltra-
Finally, considering that the inflammation is particularly deleterious in tion of inflammatory cells98. But nuclear factor-kB–dependent inflam-
ischemia associated with reperfusion91,92, suppression of inflammation matory mediators, such as IL-1, TNF, inducible nitric oxide synthase
would be a fitting complement to reperfusion therapy using thrombolyt- (iNOS)-derived NO and ROS, are also required for the full expression
ics or intravascular clot removal. of the tolerance99–102, indicating that the protection does not rely just on
Although these considerations are based on animal models, which the suppression of deleterious inflammatory mediators but also on a fine
may not recapitulate in full the human disease (Box 1), inflammation balance between pro- and anti-inflammatory signaling. One of the chal-
is a crucial pathogenic component of human stroke and remains an lenges is therefore to learn how to modulate the immune system to rep-
attractive target for therapeutic intervention. licate the beneficial inflammatory milieu induced by preconditioning.
Anti-inflammatory agents. Blocking antibodies directed against Tolerization may provide the opportunity to achieve this goal. Induction
adhesion molecules (for example, intracellular adhesion molecule-1 of immune tolerance through mucosal exposure to myelin antigens or
(ICAM-1) or macrophage-1 antigen) or recombinant neutrophil inhibi- E-selectin promotes a protective TH2 response, which acts through mul-
tory factor have not been effective in clinical trials93. In the case of the tiple pathways to suppress the deleterious effects of inflammation47. As
ICAM-1 trial, the negative outcome has been attributed to deleterious tolerization must be established before injury, this approach, like precon-
immunoactivation resulting from administration of a mouse antibody ditioning, would be more appropriate for stroke prevention in high-risk
to humans, as reproduced in an experimental study in which mouse individuals than acute stroke treatment. Another strategy may be the
antibodies to rat ICAM-1 were administered to rats94. Although there administration of RTL (Table 3), given that it suppresses the infiltra-
also might be other reasons for these failures93, a probable contribut- tion of inflammatory cells and provides neuroprotection even when
ing factor is that post-ischemic inflammation acts through multiple administered after the onset of cerebral ischemia49. Similarly, adminis-
redundant pathways that cannot be effectively suppressed by blocking tration of the immunomodulatory co-polymer Poly-YE ameliorates the
a single cytokine or adhesion molecule, as attempted in these clinical neurological deficits produced by cerebral ischemia, an effect attributed
trials. Thus, neutralizing upstream mediators of the signaling cascade to increased production of growth factors and hippocampal neurogen-
or blocking multiple inflammatory pathways would be more effective esis103. The full translational potential of treatments based on immuno-
(Table 3). For example, blocking upstream components of inflamma- modulation has not been established—therapeutic window, efficacy in
females and in aging, protection in the presence of cardiovascular risk suppression of inflammation. Furthermore, in light of stroke-induced
factors and efficacy in higher-order species need further exploration. immunosuppression, the infectious complications of therapies suppress-
Nevertheless, the powerful protective effects of immunomodulators ing inflammation also need to be taken into account (Table 3).
justify additional investigations in this direction. Therapies based on immunomodulation, in which the overall immune
The recent identification of IL-17–secreting T cells as a crucial effector response is deviated from a TH1- to a TH2-type response, also have a dark
of the tissue damage in autoimmune diseases (TH17 cells)104 and cere- side. In models of multiple sclerosis, tolerization with myelin antigens
bral ischemia (gdT cells)41 raises the possibility that counteracting IL-17 induces a protective TH2 response in the acute phase, but, in the long
could be beneficial in cerebral ischemia as it is in experimental allergic term, such TH2 response promotes B cell differentiation and leads to a
encephalomyelitis104. Boosting the protective roles of Treg cells could also humoral attack against myelin that worsens the neurological outcome106.
be beneficial42, although a destructive role of these cells has also been Worsening in the chronic phase has also been reported in tolerization
proposed105. These approaches would be desirable because they target the applied to models of cerebral ischemia107. Therefore, the delayed effects
delayed phase of the injury and are anticipated to have a particularly wide of humoral immunity could counteract the short-term benefit of suppres-
therapeutic window. Nevertheless, the role of lymphocytes in ischemic sion of cellular immunity. A more complete understanding of the immu-
injury is poorly understood, and the full implications of suppressing the nology of stroke would enable the development of targeted approaches to
action of specific T and B cell populations remain to be defined. selectively suppress the deleterious effects of inflammation.
Fighting inflammation: a double-edged sword? Immune cells
and inflammation play a key part in tissue repair and reorganization. Conclusions
These beneficial effects have to be considered in developing therapeu- Immunity and inflammation are an integral part of the pathogenic pro-
tic approaches aimed at restraining post-ischemic inflammation. The cesses triggered by ischemia and reperfusion. Inflammatory signaling
concern is that although counteracting the inflammatory response to is responsible for early molecular events triggered by the arterial occlu-
© 2011 Nature America, Inc. All rights reserved.
ischemic injury may ameliorate the tissue damage in the acute phase, sion and that culminate in the invasion of the brain by blood-borne
it may also compromise repair mechanisms and worsen the long-term leukocytes. Although its ultimate goal is to reestablish homeostasis,
outcome of the injury. Owing to the paucity of experimental studies in inflammation inflicts considerable damage to the metastable penum-
the recovery phase, there is no definitive experimental evidence that bral tissue. Adaptive immunity is deeply involved in the central and
anti-inflammatory treatments interfere with repair processes in the post- peripheral events triggered by cerebral ischemia, but whether a clas-
ischemic brain. However, prosurvival effects of immune cells stemming sical autoimmune response against brain antigens unveiled by tissue
from growth factor production, neurogenesis and neuroplasticity are damage contributes to the acute phase of the damage is still unknown.
well established (Table 2)105. The essential role of inflammation in tissue Lymphocytes invade the ischemic brain and contribute to tissue dam-
repair highlights the difficulties with approaches based on full-blown age, but the rapidity of their deleterious effect is not consistent with an
ROS and NO. In the setting of inflammation, O2– is produced Apoptosis inducing receptors. Fas (CD95), a member of the
mainly by NADPH oxidase, an enzyme expressed in virtually TNF receptor superfamily, is expressed on neurons and glia after
all inflammatory cells (Boxes 2 and 3). At the same time, large ischemia129, whereas its ligand FasL (CD95L) is present in
amounts of NO are produced by de novo expression of iNOS89. neurons, microglia cytotoxic T cells, gdT cells and NK cells137.
NO reacts preferentially with O2– to form the cytotoxic molecule Fas ligation results in formation of the death-inducing signaling
peroxynitrite. H2O2 is derived from O2– dismutation and gives complex and subsequent activation of caspase-8 and of the
rise to highly toxic hydroxyl radicals via the Haber-Weiss reaction, proapoptotic factor Bid. Similarly, the TNF-related apoptosis-
facilitated by the increased availability of free iron in ischemia123. inducing ligand (TRAIL) is expressed de novo after ischemia in
These toxic molecules alter cellular proteins, lipids and ribonucleic astrocytes and microglia138, and it induces apoptosis by engaging
acids, leading to cell dysfunction or death, and they have been its receptors on neurons and glia. This pathway is involved in
implicated in the tissue damage produced by post-ischemic ischemic cell death, because TRAIL inhibition or Fas mutation
inflammation. For example, suppression of NADPH oxidase or ameliorates ischemic injury138,139.
iNOS activity protects the brain in the late phase of cerebral
ischemia (Supplementary Table 1). Perforin and granzyme. This pathway is used by cytotoxic T cells,
NKT and NK cells, which release perforin and granzyme upon
The complement system. The complement system is a proteolytic molecular recognition of antigen associated with MHC class I or
cascade composed of several subunits (C1 through C9) that class I–like molecules. After engagement of accessory surface
leads to cell lysis via the formation of a pore-like structure receptors, such as ICAM-1, degranulation occurs releasing
assembled from C9 termed the membrane attack complex. the protease granzyme, perforin, which is analogous to the
Complement also enhances phagocytosis (opsonization), or acts complement subunit C9, and the proteoglycan serglycin. This
as a chemotactic-activating stimulus for inflammatory cells. After complex is internalized by the target cell, where granzyme triggers
ischemia, complement is activated, and its components are either apoptosis by activating caspase-3 and Bid. The involvement of this
upregulated in glia and neurons or enter the brain through BBB pathway in ischemic brain injury is suggested by observations that
breakdown136. The involvement of complement in ischemic damage genetic deletion of perforin is protective140.
adaptive immune response targeted to the brain. Remarkably, selected leukocyte surface apyrase CD39. J. Clin. Invest. 119, 1136–1149 (2009).
12. Yilmaz, G. & Granger, D.N. Leukocyte recruitment and ischemic brain injury.
lymphocyte subpopulations are protective, acting to dampen the cyto- Neuromolecular Med. 12, 193–204 (2010).
toxic effects of other inflammatory cells and promoting tissue recovery. 13. Atochin, D.N. et al. The phosphorylation state of eNOS modulates vascular reac-
The mechanistic bases for this dichotomous response of lymphocytes tivity and outcome of cerebral ischemia in vivo. J. Clin. Invest. 117, 1961–1967
(2007).
remains to be elucidated (Fig. 3). Although the participation of inflam- 14. Ishikawa, M., Zhang, J.H., Nanda, A. & Granger, D.N. Inflammatory responses to isch-
mation and immunity in tissue recovery is well established in other emia and reperfusion in the cerebral microcirculation. Front. Biosci. 9, 1339–1347
organs, very little is known about these processes in the brain. Even (2004).
15. Yemisci, M. et al. Pericyte contraction induced by oxidative-nitrative stress impairs
less clear are the long-term effects of the adaptive immune response capillary reflow despite successful opening of an occluded cerebral artery. Nat. Med.
associated with stroke and their role in the sequelae of ischemic dam- 15, 1031–1037 (2009).
16. Engelhardt, B. & Sorokin, L. The blood-brain and the blood-cerebrospinal fluid bar-
age, such as brain atrophy and dementia. Similarly, it is unclear whether riers: function and dysfunction. Semin. Immunopathol. 31, 497–511 (2009).
the immune system develops a memory of the antigen exposure and 17. Lindsberg, P.J., Strbian, D. & Karjalainen-Lindsberg, M.L. Mast cells as early respond-
whether an autoimmune response develops after subsequent exposure to ers in the regulation of acute blood-brain barrier changes after cerebral ischemia and
hemorrhage. J. Cereb. Blood Flow Metab. 30, 689–702 (2010).
the same antigen, as it may occur in cases of recurrent strokes. Although 18. Strbian, D., Karjalainen-Lindsberg, M.L., Tatlisumak, T. & Lindsberg, P.J. Cerebral
much has been learned on the interplay between the peripheral immu- mast cells regulate early ischemic brain swelling and neutrophil accumulation.
nosuppression and the central immune activation associated with stroke, J. Cereb. Blood Flow Metab. 26, 605–612 (2006).
19. Konsman, J.P., Drukarch, B. & Van Dam, A.M. (Peri)vascular production and action
their impact on short- and long-term tissue outcome remains poorly of pro-inflammatory cytokines in brain pathology. Clin. Sci. (Lond.) 112, 1–25
understood. The fact that the immune system and inflammation are (2007).
20. Sairanen, T.R., Lindsberg, P.J., Brenner, M. & Siren, A.L. Global forebrain ischemia
central to the pathophysiology of stroke has raised the prospect of new results in differential cellular expression of interleukin-1b (IL-1b) and its receptor
therapeutic approaches to counteract ischemic injury. However, under- at mRNA and protein level. J. Cereb. Blood Flow Metab. 17, 1107–1120 (1997).
standing of the crosstalk between the immune system and the ischemic 21. Kono, H. & Rock, K.L. How dying cells alert the immune system to danger. Nat. Rev.
© 2011 Nature America, Inc. All rights reserved.
45. Frenkel, D., et al. Nasal vaccination with myelin oligodendrocyte glycoprotein reduces on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain Res. 955,
stroke size by inducing IL-10–producing CD4+ T cells. J. Immunol. 171, 6549–6555 85–97 (2002).
(2003). 80. Vogelgesang, A. et al. Functional status of peripheral blood T-cells in ischemic stroke
46. Becker, K., Kindrick, D., McCarron, R., Hallenbeck, J. & Winn, R. Adoptive transfer patients. PLoS ONE 5, e8718 (2010).
of myelin basic protein-tolerized splenocytes to naive animals reduces infarct size: 81. Chamorro, A., Urra, X. & Planas, A.M. Infection after acute ischemic stroke: a mani-
a role for lymphocytes in ischemic brain injury? Stroke 34, 1809–1815 (2003). festation of brain-induced immunodepression. Stroke 38, 1097–1103 (2007).
47. Becker, K.J. Sensitization and tolerization to brain antigens in stroke. Neuroscience 82. Prass, K. et al. Stroke-induced immunodeficiency promotes spontaneous bacterial
158, 1090–1097 (2009). infections and is mediated by sympathetic activation reversal by poststroke T helper
48. Frenkel, D. et al. Neuroprotection by IL-10–producing MOG CD4+ T cells following cell type 1-like immunostimulation. J. Exp. Med. 198, 725–736 (2003).
ischemic stroke. J. Neurol. Sci. 233, 125–132 (2005). 83. Leys, D., Henon, H., Mackowiak-Cordoliani, M.A. & Pasquier, F. Poststroke dementia.
49. Subramanian, S. et al. Recombinant T cell receptor ligand treats experimental stroke. Lancet Neurol. 4, 752–759 (2005).
Stroke 40, 2539–2545 (2009). 84. McColl, B.W., Rothwell, N.J. & Allan, S.M. Systemic inflammatory stimulus poten-
50. Itakura, A. et al. Characterization of human platelet binding of recombinant T cell tiates the acute phase and CXC chemokine responses to experimental stroke and
receptor ligand. J. Neuroinflammation 7, 75 (2010). exacerbates brain damage via interleukin-1– and neutrophil-dependent mechanisms.
51. Abbas, A.K. Basic Immunology Updated Edition: Functions and Disorders of the J. Neurosci. 27, 4403–4412 (2007).
Immune System Ch. 5 (Saunders, 2010). 85. Becker, K.J., Kindrick, D.L., Lester, M.P., Shea, C. & Ye, Z.C. Sensitization to brain
52. Yilmaz, G., Arumugam, T.V., Stokes, K.Y. & Granger, D.N. Role of T lymphocytes and antigens after stroke is augmented by lipopolysaccharide. J. Cereb. Blood Flow
interferon-g in ischemic stroke. Circulation 113, 2105–2112 (2006). Metab. 25, 1634–1644 (2005).
53. Offner, H., Vandenbark, A.A. & Hurn, P.D. Effect of experimental stroke on peripheral 86. Zierath, D. et al. CNS immune responses following experimental stroke. Neurocrit.
immunity: CNS ischemia induces profound immunosuppression. Neuroscience 158, Care 12, 274–284 (2010).
1098–1111 (2009). 87. Fonarow, G.C. et al. Timeliness of tissue-type plasminogen activator therapy in acute
54. Andrews, D.M. & Smyth, M.J. A potential role for RAG-1 in NK cell development ischemic stroke: patient characteristics, hospital factors and outcomes associated
revealed by analysis of NK cells during ontogeny. Immunol. Cell Biol. 88, 107–116 with door-to-needle times within 60 minutes. Circulation 123, 750–758 (2011).
(2010). 88. Gesuete, R. et al. Recombinant C1 inhibitor in brain ischemic injury. Ann. Neurol.
55. Spite, M. & Serhan, C.N. Novel lipid mediators promote resolution of acute inflam- 66, 332–342 (2009).
mation: impact of aspirin and statins. Circ. Res. 107, 1170–1184 (2010). 89. Iadecola, C., Zhang, F. & Xu, X. Inhibition of inducible nitric oxide synthase amelio-
56. Nathan, C. & Ding, A. Nonresolving inflammation. Cell 140, 871–882 (2010). rates cerebral ischemic damage. Am. J. Physiol. 268, R286–R292 (1995).
© 2011 Nature America, Inc. All rights reserved.
57. Schilling, M. et al. Predominant phagocytic activity of resident microglia over 90. Lee, S.-T., et al. Anti-inflammatory mechanism of intravascular neural stem cell
hematogenous macrophages following transient focal cerebral ischemia: an investi- transplantation in haemorrhagic stroke. Brain 131, 616–629 (2008).
gation using green fluorescent protein transgenic bone marrow chimeric mice. Exp. 91. Prestigiacomo, C.J. et al. CD18-mediated neutrophil recruitment contributes to the
Neurol. 196, 290–297 (2005). pathogenesis of reperfused but not nonreperfused stroke. Stroke 30, 1110–1117
58. Denes, A. et al. Proliferating resident microglia after focal cerebral ischaemia in mice. (1999).
J. Cereb. Blood Flow Metab. 27, 1941–1953 (2007). 92. Stowe, A.M. et al. Neutrophil elastase and neurovascular injury following focal stroke
59. Rappert, A. et al. CXCR3-dependent microglial recruitment is essential for dendrite and reperfusion. Neurobiol. Dis. 35, 82–90 (2009).
loss after brain lesion. J. Neurosci. 24, 8500–8509 (2004). 93. del Zoppo, G.J. Lessons from stroke trials using anti-inflammatory approaches that
60. Davalos, D. et al. ATP mediates rapid microglial response to local brain injury in vivo. have failed. in Neuroinflammation in Stroke (eds. Dirnagl, U. & Elger, B.) 155–184
Nat. Neurosci. 8, 752–758 (2005). (Springer-Verlagh, Berlin–Heidelberg, 2004).
61. Taylor, A., Verhagen, J., Blaser, K., Akdis, M. & Akdis, C.A. Mechanisms of immune 94. Furuya, K. et al. Examination of several potential mechanisms for the negative out-
suppression by interleukin-10 and transforming growth factor-b: the role of T regula- come in a clinical stroke trial of enlimomab, a murine anti-human intercellular
tory cells. Immunology 117, 433–442 (2006). adhesion molecule-1 antibody: a bedside-to-bench study. Stroke 32, 2665–2674
62. Greenberg, D.A. & Jin, K. Growth factors and stroke. NeuroRx 3, 458–465 (2006). (2001).
63. Carmichael, S.T. Translating the frontiers of brain repair to treatments: starting not 95. Fagan, S.C. et al. Minocycline to improve neurologic outcome in stroke (MINOS): a
to break the rules. Neurobiol. Dis. 37, 237–242 (2010). dose-finding study. Stroke 41, 2283–2287 (2010).
64. Zhang, Z.G., Zhang, L., Jiang, Q. & Chopp, M. Bone marrow–derived endothelial pro- 96. Jensen, H.A. et al. Remote ischemic preconditioning protects the brain against injury
genitor cells participate in cerebral neovascularization after focal cerebral ischemia after hypothermic circulatory arrest. Circulation 123, 714–721 (2011).
in the adult mouse. Circ. Res. 90, 284–288 (2002). 97. Karikó, K., Weissman, D. & Welsh, F.A. Inhibition of Toll-like receptor and cytokine
65. Hayakawa, K., Qiu, J. & Lo, E.H. Biphasic actions of HMGB1 signaling in inflamma- signaling–a unifying theme in ischemic tolerance. J. Cereb. Blood Flow Metab. 24,
tion and recovery after stroke. Ann. NY Acad. Sci. 1207, 50–57 (2010). 1288–1304 (2004).
66. Li, S. et al. An age-related sprouting transcriptome provides molecular control of 98. Marsh, B. et al. Systemic lipopolysaccharide protects the brain from ischemic
axonal sprouting after stroke. Nat. Neurosci. 13, 1496–1504 (2010). injury by reprogramming the response of the brain to stroke: a critical role for IRF3.
67. Lalancette-Hébert, M., Gowing, G., Simard, A., Weng, Y.C. & Kriz, J. Selective J. Neurosci. 29, 9839–9849 (2009).
ablation of proliferating microglial cells exacerbates ischemic injury in the brain. 99. Blondeau, N., Widmann, C., Lazdunski, M. & Heurteaux, C. Activation of the nuclear
J. Neurosci. 27, 2596–2605 (2007). factor-kB is a key event in brain tolerance. J. Neurosci. 21, 4668–4677 (2001).
68. Hayakawa, K., et al. Inhibition of reactive astrocytes with fluorocitrate retards neu- 100. Kunz, A. et al. Neurovascular protection by ischemic tolerance: role of nitric oxide
rovascular remodeling and recovery after focal cerebral ischemia in mice. J. Cereb. and reactive oxygen species. J. Neurosci. 27, 7083–7093 (2007).
Blood Flow Metab. 30, 871–882 (2010). 101. Ohtsuki, T., Ruetzler, C.A., Tasaki, K. & Hallenbeck, J.M. Interleukin-1 mediates
69. Zhang, Z.G. et al. Correlation of VEGF and angiopoietin expression with disruption of induction of tolerance to global ischemia in gerbil hippocampal CA1 neurons.
blood-brain barrier and angiogenesis after focal cerebral ischemia. J. Cereb. Blood J. Cereb. Blood Flow Metab. 16, 1137–1142 (1996).
Flow Metab. 22, 379–392 (2002). 102. Pradillo, J.M., et al. TNFR1 upregulation mediates tolerance after brain ischemic
70. Hao, Q., et al. Neutrophil depletion decreases VEGF-induced focal angiogenesis in preconditioning. J. Cereb. Blood Flow Metab. 25, 193–203 (2005).
the mature mouse brain. J. Cereb. Blood Flow Metab. 27, 1853–1860 (2007). 103. Ziv, Y. et al. A novel immune-based therapy for stroke induces neuroprotection and
71. Manoonkitiwongsa, P.S., Schultz, R.L., McCreery, D.B., Whitter, E.F. & Lyden, P.D. supports neurogenesis. Stroke 38, 774–782 (2007).
Neuroprotection of ischemic brain by vascular endothelial growth factor is critically 104. Steinman, L. A brief history of TH17, the first major revision in the TH1/TH2 hypoth-
dependent on proper dosage and may be compromised by angiogenesis. J. Cereb. esis of T cell–mediated tissue damage. Nat. Med. 13, 139–145 (2007).
Blood Flow Metab. 24, 693–702 (2004). 105. Schwartz, M., London, A. & Shechter, R. Boosting T-cell immunity as a therapeutic
72. Zhang, Z.G. et al. VEGF enhances angiogenesis and promotes blood-brain barrier approach for neurodegenerative conditions: the role of innate immunity. Neuroscience
leakage in the ischemic brain. J. Clin. Invest. 106, 829–838 (2000). 158, 1133–1142 (2009).
73. Denes, A. et al. Experimental stroke-induced changes in the bone marrow reveal com- 106. Genain, C.P. et al. Late complications of immune deviation therapy in a nonhuman
plex regulation of leukocyte responses. J. Cereb. Blood Flow Metab. 31, 1036–1050 primate. Science 274, 2054–2057 (1996).
(2011). 107. Gee, J.M. et al. Long term immunologic consequences of experimental stroke and
74. Jickling, G.C. et al. Signatures of cardioembolic and large-vessel ischemic stroke. mucosal tolerance. Exp. Transl. Stroke Med. 1, 3 (2009).
Ann. Neurol. 68, 681–692 (2010). 108. Schroder, K. & Tschopp, J. The inflammasomes. Cell 140, 821–832 (2010).
75. Offner, H. et al. Experimental stroke induces massive, rapid activation of the periph- 109. Kunz, A. et al. Nuclear factor-kB activation and postischemic inflammation are sup-
eral immune system. J. Cereb. Blood Flow Metab. 26, 654–665 (2006). pressed in CD36-null mice after middle cerebral artery occlusion. J. Neurosci. 28,
76. Emsley, H.C.A. et al. An early and sustained peripheral inflammatory response 1649–1658 (2008).
in acute ischaemic stroke: relationships with infection and atherosclerosis. 110. Arumugam, T.V., Granger, D.N. & Mattson, M.P. Stroke and T-cells. Neuromolecular
J. Neuroimmunol. 139, 93–101 (2003). Med. 7, 229–242 (2005).
77. Smith, C.J. et al. Peak plasma interleukin-6 and other peripheral markers of inflam- 111. Brait, V.H. et al. Mechanisms contributing to cerebral infarct size after stroke: gen-
mation in the first week of ischaemic stroke correlate with brain infarct volume, stroke der, reperfusion, T lymphocytes and Nox2-derived superoxide. J. Cereb. Blood Flow
severity and long-term outcome. BMC Neurol. 4, 2 (2004). Metab. 30, 1306–1317 (2010).
78. Haeusler, K.G. et al. Immune responses after acute ischemic stroke or myocardial 112. Napoli, I. & Neumann, H. Microglial clearance function in health and disease.
infarction. Int. J. Cardiol. published online, doi:10.1016/j.ijcard.2010.10.053 Neuroscience 158, 1030–1038 (2009).
(13 November 2010). 113. Hanayama, R. et al. Autoimmune disease and impaired uptake of apoptotic cells in
79. Gendron, A. et al. Temporal effects of left versus right middle cerebral artery occlusion MFG-E8-deficient mice. Science 304, 1147–1150 (2004).
114. Buisson, A. et al. Up-regulation of a serine protease inhibitor in astrocytes medi- blood-borne macrophages. Eur. J. Neurosci. 14, 1651–1658 (2001).
ates the neuroprotective activity of transforming growth factor b1. FASEB J. 12, 127. Mantovani, A., Sica, A. & Locati, M. Macrophage polarization comes of age. Immunity
1683–1691 (1998). 23, 344–346 (2005).
115. Grilli, M. et al. Interleukin-10 modulates neuronal threshold of vulnerability to ischae- 128. Rao, K.N. & Brown, M.A. Mast cells: multifaceted immune cells with diverse roles in
mic damage. Eur. J. Neurosci. 12, 2265–2272 (2000). health and disease. Ann. NY Acad. Sci. 1143, 83–104 (2008).
116. Arai, K., Jin, G., Navaratna, D. & Lo, E.H. Brain angiogenesis in developmental and 129. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 33, 657–670
pathological processes: neurovascular injury and angiogenic recovery after stroke. (2010).
FEBS J. 276, 4644–4652 (2009). 130. Mestas, J. & Hughes, C.C. Of mice and not men: differences between mouse and
117. Jauch, E.C. et al. Association of serial biochemical markers with acute ischemic human immunology. J. Immunol. 172, 2731–2738 (2004).
stroke: the National Institute of Neurological Disorders and Stroke recombinant 131. Kreisel, D. et al. Mouse vascular endothelium activates CD8+ T lymphocytes in a
tissue plasminogen activator Stroke Study. Stroke 37, 2508–2513 (2006). B7-dependent fashion. J. Immunol. 169, 6154–6161 (2002).
118. Bornstein, N.M. et al. Antibodies to brain antigens following stroke. Neurology 56, 132. Wan, Y.Y. Multi-tasking of helper T cells. Immunology 130, 166–171 (2010).
529–530 (2001). 133. Biron, C.A., Nguyen, K.B., Pien, G.C., Cousens, L.P. & Salazar-Mather, T.P. Natural
119. Dambinova, S.A. et al. Blood test detecting autoantibodies to N-methyl-d-aspartate killer cells in antiviral defense: function and regulation by innate cytokines. Annu. Rev.
neuroreceptors for evaluation of patients with transient ischemic attack and stroke. Immunol. 17, 189–220 (1999).
Clin. Chem. 49, 1752–1762 (2003). 134. Bonneville, M., O’Brien, R.L. & Born, W.K. gd T cell effector functions: a blend of innate
120. Rocklin, R.E., Sheremata, W.A., Feldman, R.G., Kies, M.W. & David, J.R. The programming and acquired plasticity. Nat. Rev. Immunol. 10, 467–478 (2010).
Guillain-Barré syndrome and multiple sclerosis. In vitro cellular responses to nervous- 135. Hayday, A.C. gd T cells and the lymphoid stress-surveillance response. Immunity 31,
tissue antigens. N. Engl. J. Med. 284, 803–808 (1971). 184–196 (2009).
121. Gordon, P.H. et al. Efficacy of minocycline in patients with amyotrophic lateral 136. Yanamadala, V. & Friedlander, R.M. Complement in neuroprotection and neurodegen-
sclerosis: a phase III randomised trial. Lancet Neurol. 6, 1045–1053 (2007). eration. Trends Mol. Med. 16, 69–76 (2010).
122. Koch, S., Katsnelson, M., Dong, C. & Perez-Pinzon, M. Remote ischemic limb precon- 137. Strasser, A., Jost, P.J. & Nagata, S. The many roles of FAS receptor signaling in the
ditioning after subarachnoid hemorrhage: a phase Ib study of safety and feasibility. immune system. Immunity 30, 180–192 (2009).
Stroke 42, 1387–1391 (2011). 138. Cui, M. et al. Blocking TRAIL-DR5 signaling with soluble DR5 reduces delayed neu-
123. Lipton, P. Ischemic cell death in brain neurons. Physiol. Rev. 79, 1431–1568 ronal damage after transient global cerebral ischemia. Neurobiol. Dis. 39, 138–147
(1999). (2010).
124. Bacigaluppi, M., Comi, G. & Hermann, D.M. Animal models of ischemic stroke. Part 139. Martin-Villalba, A. et al. CD95 ligand (Fas-L/APO-1L) and tumor necrosis factor-related
© 2011 Nature America, Inc. All rights reserved.
one: modeling risk factors. Open Neurol. J. 4, 26–33 (2010). apoptosis-inducing ligand mediate ischemia-induced apoptosis in neurons. J. Neurosci.
125. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from 19, 3809–3817 (1999).
primitive macrophages. Science 330, 841–845 (2010). 140. Liesz, A. et al. Inhibition of lymphocyte trafficking shields the brain against deleterious
126. Bechmann, I. et al. Immune surveillance of mouse brain perivascular spaces by neuroinflammation after stroke. Brain 134, 704–720 (2011).