Sunteți pe pagina 1din 14

NeuroMolecular Medicine

Copyright 2005 Humana Press Inc.


All rights of any nature whatsoever reserved.
ISSN1535-1084/05/07:229242/$30.00
doi: 10.1385/NMM:7:3:229

REVIEW ARTICLE

Stroke and T-Cells

Thiruma V. Arumugam,*,1 D. Neil Granger,2 and Mark P. Mattson1


1
Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, 5600 Nathan
Shock Drive, Baltimore, MD 21224; and 2Department of Molecular and Cellular Physiology, Louisiana
State University Health Sciences Center, Shreveport, LA 71130
Received August 10, 2005; Revised August 11, 2005; Accepted August 11, 2005

Abstract
The microvasculature of the brain region affected by a stroke assumes an inflammatory phenotype
that is characterized by endothelial cell activation and barrier dysfunction and the recruitment of
adherent leukocytes. Although most attention has been devoted to the possible role of neutrophils
in the tissue responses to ischemic stroke there is evidence that T-lymphocytes also accumulate in
the postischemic brain. Although comparable detailed analyses of lymphocyte involvement in
ischemic brain injury have not been performed, emerging findings suggest a role for T-cells in the
pathogenesis of ischemic stroke. The recruitment of T-cells to the site of brain injury is critically
dependent on the coordinated expression of adhesion molecules on the activated capillary endothe-
lium. Whether the recruited lymphocytes are acting directly on brain tissue or indirectly through
activation of other circulating blood cells and/or extravascular cells remain unclear. Cytotoxic
CD8+ T-cells may induce brain injury through molecules released from their cytotoxic granules.
CD4+ T-helper 1 (TH1) cells, which secrete proinflammatory cytokines, including interleukin-2 (IL-
2), IL-12, interferon-, and tumor necrosis factor-, may play a key role in the pathogenesis of stroke,
whereas CD4+ TH2 cells may play a protective role through anti-inflammatory cytokines such as
IL-4, IL-5, IL-10, and IL-13. T-cells should be considered as therapeutic targets for ischemic stroke.
However, because infection is a leading cause of mortality in the postacute phase of ischemic stroke,
and considering anti-inflammatory role of CD4+ TH2, treatment targeting T-cells should be carefully
designed to reduce deleterious and enhance protective actions of T-cells.
doi: 10.1385/NMM:7:3:229
Index Entries: Leukocyte; adhesion; stroke; T-cells; ischemia/reperfusion injuries; inflammation.

Introduction stroke and limiting ischemic damage are major goals


to decrease stroke mobility and mortality. Brain
Stroke is a leading cause of morbidity and mor- tissue is exquisitely sensitive to oxygen and glucose
tality in the world, resulting in the death of approx deprivation such that even brief ischemia can initi-
(5) 106 people per year. Enhancing recovery from ate a complex sequence of events that ultimately
*Author to whom all correspondence and reprint requests should be addressed. E-mail: arumugamt@mail.nih.gov

NeuroMolecular Medicine 229 Volume 7, 2005


230 Arumugam et al.

culminates in cellular death. The pathophysiologi- to this pathological condition. Most attention has
cal processes in stroke are complex and involve been devoted to the possible role of neutrophils
disruption of the bloodbrain barrier (BBB), energy because this leukocyte subset accumulates early
failure, loss of cell ion homeostasis, acidosis, after reperfusion. However, there is evidence that
increased intracellular calcium levels, excitotoxicity, T-lymphocytes also accumulate in the postischemic
free radical-mediated toxicity, generation of arachi- brain within a few hours after reperfusion. Cytotoxic
donic acid products, cytokine-mediated apoptosis, T-cells can contribute to tissue damage by several
activation of glial cells, and infiltration of leuko- mechanisms including release of inflammatory
cytes. Evidence of an inflammatory component in cytokines that recruit other leukocyte subsets.
stroke comes from the observations that ischemic Although there is a limited amount of evidence
injury can be attenuated by preischemic induction supporting a role for T-lymphocytes in ischemic
of systemic neutropenia (Ishikawa et al., 2004a), brain injury, there is a large body of evidence that
pharmacological blockage of adhesion molecules or invokes a major role for lymphocytes in the patho-
their receptors (DeGraba, 1998; Frijns and Kappelle, genesis of I/R injury in other vascular beds (Le
2002), targeted disruption of genes encoding inflam- Moine et al., 2000; Shigematsu et al., 2002; Burne-
matory cytokines, and interfering with the action Taney et al., 2003). This article will piece together
of inflammatory mediators (Dirnagl et al., 2003; Han recent findings related to T-cell involvement in
and Yenari, 2003). Recent studies reveal that brain stroke injury pathogenesis.
ischemia is a powerful stimulus to trigger a series
of events that lead to vasodilatation and increased
permeability of local blood vessels, and mobiliza- Evidence for T-Cell Involvement
tion and infiltration of circulating leukocytes, a in Stroke
common process shared by all ischemia/reperfu-
sion (I/R) injuries. Immunohistochemical and flow cytometric stud-
In response to I/R, the microvasculature assumes ies of postischemic brain tissue have demonstrated
an inflammatory phenotype characterized by leuko- that the initial rapid recruitment of neutrophils is
cyteendothelial cell adhesion, leukocyte capillary followed over the subsequent hours to days by the
plugging, endothelial barrier dysfunction, activa- accumulation of other leukocyte subsets, including
tion of resident macrophages, and an enhanced gen- lymphocytes and monocytes (Campanella et al.,
eration of oxidants and inflammatory mediators 2002; Stevens et al., 2002). Intravital microscopic
(Ishikawa et al., 2004b). The mechanisms that studies of the postischemic brain microcirculation
underlie the microvascular dysfunction and brain have also revealed that the accumulation of leuko-
cell death that are associated with ischemic stroke cytes in postcapillary venules is generally accom-
are currently an area of intense investigation. panied by the recruitment of adherent platelets,
Indeed, there are several lines of evidence that sup- suggesting that the venules assume both a proin-
port a role for leukocyte-mediated inflammation in flammatory and prothrombogenic phenotype after
the pathogenesis of ischemic stroke. The contention I/R (Ishikawa et al., 2003; Arumugam et al., 2004).
that leukocyte-mediated inflammation is a cause Although the impact of the accumulated platelets
rather than merely an outcome of the brain injury in the postischemic microcirculation and recruit-
resulting from I/R is supported by reports that ment of T-lymphocytes is not known, these cells
administration of blocking antibodies against spe- may enhance thrombosis and/or neurovascular
cific cell adhesion molecules (CAMs) that mediate damage with hemorrhage. The binding of activated
leukocyte recruitment can reduce infarct size and brain platelets to endothelial cells and/or leukocytes
edema in animal models of stroke (Matsuo et al., 1994; appears to be important during inflammation
Prestigiacomo et al., 1999). Although leukocyte because it can influence the intensity of an inflam-
recruitment and activation are strongly implicated in matory response through the release of different
the microvascular dysfunction and tissue injury asso- bioactive compounds. In our intravital microscopy
ciated with ischemic stroke, relatively little is known study of leukocyte adhesion in postischemic cere-
about the contribution of specific leukocyte subpop- bral venules, treatment of mice with antineutrophil
ulations such as CD8+ and CD4+ T-lymphocytes serum to render them completely neutropenic only

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 231

reduced the number of adherent leukocytes by 60%, permanent MCAO, there is also a twofold larger
suggesting that leukocyte subsets in addition to neu- number of T-lymphocytes in the ischemic vs non-
trophils are trafficking through the brain microcir- ischemic hemispheres (Campanella et al., 2002). The
culation 4 h after reperfusion (Ishikawa et al., 2004a). immunosuppressive agent FK506, which is used to
The revelation that T-lymphocytes participate in the prevent T-cell-mediated allograft rejection as well
early neutrophil recruitment and tissue damage fol- as other autoimmune diseases, significantly reduced
lowing ischemic injury in other organs raises the brain infarct size in a rat model of focal cerebral artery
question of whether T-cells also contribute to stroke- occlusion (Brecht et al., 2003). However, another
induced microvascular dysfunction and tissue widely used immunosuppressive agent, cyclosporine,
damage. as well as the T-cell activation inhibitor rapamycin,
The role for lymphocytes in the development of did not afford protection in the same model system
ischemic injury is supported by reports describing (Bochelen et al., 1999; Phillis et al., 2002).
significantly blunted injury responses in mice that
were genetically deficient in one or more lympho-
cyte subpopulations. It has been reported that CD8+- Mechanism of T-Cell Recruitment
or CD4+-deficient mice exhibit an improved kidney in Stroke
function after renal I/R (Ysebaert et al., 2004; Burne-
Taney et al., 2005) and reduced hepatocellular injury There are several potential mechanisms/path-
after liver I/R (Zwacka et al., 1997). There are sev- ways that T-cells can employ to promote the adhe-
eral reports describing a critical role for different sion of lymphocytes, neutrophils, and platelets, and
lymphocyte-specific (CD11a, CD11b/CD18) and the subsequent tissue injury/organ dysfunction fol-
endothelial cell (intercellular adhesion molecule lowing ischemic stroke (see Fig. 1). Under quiescent
[ICAM-1], P-selectin) adhesion molecules in medi- conditions, neutrophils circulate freely and do not
ating the brain edema and necrosis, and resultant interact significantly with the endothelium. In con-
neurological deficit, caused by transient cerebral trast, monocytes and lymphocytes exhibit a
ischemia (Prestigiacomo et al., 1999; Arumugam continuous, low-level, physiological traffic across
et al., 2004; Vemuganti et al., 2004). All of the above- the vessel wall. Monocytes emigrate from the blood-
listed criteria used to implicate a role for adherent stream to mature into tissue macrophages (microglia
T-cells in the pathogenesis of I/R injury have been in the brain) that may develop tissue- or organ-spe-
met in animal models of cerebral ischemia. In addi- cific functions (Guo et al., 2004). Immune surveil-
tion, evidence derived from studies employing lance of tissue requires that lymphocytes recirculate
immunosuppressive agents supported the involve- between blood and lymph nodes, gaining entrance
ment of these immune cells in the pathogenesis of to the latter at the high endothelial venules of post-
ischemic stroke. Together with granulocytes, many capillary venules in lymphoid tissue. Studies using
T-cells identified by immuno-cytochemistry intravital video microscopy have established a
invaded the infarct region (Schroeter et al., 1994; sequence of events involved in leukocyte emigra-
Jander et al., 1995). After both permanent middle tion to extravascular sites of inflammation (Ishikawa
cerebral artery occlusion (MCAO) and photochem- et al., 2004b).
ically induced ischemia, CD4+ T-cells infiltrated the The initial, rapidly reversible adhesion of leuko-
infarct region from day 1 onward. At day 3, their cytes to the endothelium under conditions of flow
number increased further and reached a peak produces rolling, which is the consequence of shear
around day 7, followed by a substantial decrease forces acting on the leukocyte and adhesive inter-
within the next 7 d. T-cells accumulated in the actions between selectin receptors and their glycol-
boundary zone of the infarction, often in close vicin- conjugate counterstructures. Rolling is mediated
ity to blood vessels. Flow-cytometric analyses of the primarily by the interaction of E- and P-selectin on
inflammatory cells that accumulated in the ischemic activated endothelial cells and leukocytes
rat brain (using specific surface markers for differ- L-selectins with sialylated and fucosylated carbo-
ent leukocyte subsets) indicated that although there hydrate moieties such as those expressed on various
is a fourfold larger number of neutrophils in ischemic membrane glycoproteins, particularly P-selectin
vs nonischemic hemispheres at 24 h following glycoprotein ligand-1 (PSGL-1) (Tailor and Granger,

NeuroMolecular Medicine Volume 7, 2005


232 Arumugam et al.

Fig. 1. Mechanisms of T-cell involvement in stroke.

2000). Rolling is initiated primarily by activation of increase adhesivity rapidly. Once lymphocytes are
the endothelium by extravascular stimuli such as tethered to the endothelium by selectin interactions,
bacteria-derived products or by endogenous medi- the leukocyte integrin receptors are activated by
ators produced by the endothelium or tissue cells. endothelial membrane-expressed platelet-activating
Early on, rolling is mediated by endothelial factor (PAF), endothelial membrane-bound
P-selectin and leukocyte L-selectin. E-selectin is chemokines, or locally secreted chemoattractants
involved only at later time-points, as it is not con- (Hogg et al., 2004). Activation of the leukocyte
stitutively expressed or mobilized but rather is integrins increases their affinity for their endothe-
induced over hours by de novo synthesis. Leukocyte lial ligands of the immunoglobulin gene super-
integrin receptors must be minimally adhesive in family (IgSF). The leukocyte integrins involved in
order for them to circulate freely, but are able to adhesion to endothelial cells include lymphocyte

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 233

function antigen-1, (LFA-1 or CD11a/CD18), and reports describing the presence of activated
macrophage antigen-1 (Mac-1 or CD11b/CD18), and circulating T-lymphocytes in blood within the first
very late antigen-4 (VLA-4, CD49d/CD29). The IgSF 9 h of reperfusion following stroke in humans
ligands are ICAM-1 and ICAM-2 for the 2-integrins, (Garlichs et al., 2003).
and vascular CAM (VCAM)-1 for VLA-4. The pres- The recognition that leukocyteendothelial cell
ence of recruited T-cells at the site of inflammation adhesion is an important component of the injury
is critically dependent on the coordinated expres- process in postischemic brain has resulted in an
sion of these adhesion molecules and the activated effort to characterize the kinetics of leukocyte accu-
capillary endothelium. mulation in capillaries (plugging) and postcapillary
Naive T-cells that are not activated generally do venules (adhesion) after I/R and to define the spe-
not cross the BBB. T-cells readily cross the BBB if a cific CAMs that mediate this recruitment process.
T-cell response against a central nervous system The leukocyte recruitment response to brain injury
(CNS) autoantigen is initiated in lymphoid organs. has been studied systematically following acute
T-cells are restimulated on an encounter with the focal ischemia by several laboratories. Many com-
target immunogen presented by local antigen- pounds released by activated neutrophils and
presenting cells (APCs). Thus, several types of CNS endothelial cells may act as platelet agonists (e.g.,
cells that may act as APCs are activated following superoxide, hydrogen peroxide) or antagonists
stroke, including microglia, and produce proin- (ADPases); conversely, platelets can release factors
flammatory cytokines such as tumor necrosis factor that may either inhibit or activate neutrophils (Ward
(TNF)-, interleukin (IL)-1, and IL-6, which may et al., 1988; Flad et al., 1997; Carden and Granger,
enhance the appearance of adhesion molecules 2000). Platelets can also facilitate the formation of
(Price et al., 2003). These cells could act to enhance inflammatory mediators by endothelial cells
the recruitment of T-cells. One possibility is that the through transcellular exchange of precursor
T-cells adhere to venular endothelium, enter the metabolites (Whatley et al., 1990). Furthermore,
tissue, and directly elicit cell necrosis. This possi- platelets can enhance the recruitment of leukocytes
bility is supported by reports describing the pres- into inflamed tissue by serving as a P-selectin-rich
ence of T-cells in the brain early after reperfusion. platform on endothelium for leukocyte adhesion
T-cell recruitment into the CNS is usually observed (Vandendries et al., 2004) and by reducing shear
in autoimmune and inflammatory diseases such as rates in the microcirculation through the release of
experimental autoimmune encephalomyelitis potent vasoconstrictors (e.g., thromboxane A).
(EAE) (Stoll et al., 1993; Chavarria and Alcocer- Similarly, although several intravital microscopic
Varela, 2004). In EAE, systemic immunization with studies have attempted to define the molecular
myelin proteins such as myelin basic protein (MBP) determinants of the adhesion of the general leukocyte
creates CD4+ helper T-cells that are antigen-specific population in the postischemic cerebral microcircu-
and enter the CNS after 1012 d. Second, large num- lation, there have been no attempts to define the
bers of nonspecific T-cells and macrophages infil- adhesion molecules that account specifically for the
trate the CNS and cause myelin destruction and enhanced lymphocyte trafficking (T-cells) in the brain
clinical disease. In cerebral ischemia, the period microcirculation after transient focal ischemia. Such
between lesion induction and T-cell infiltration is molecular mechanisms of lymphocyte recruitment
too short for generation of a systemic, antigen-spe- have been assessed, however, in brain microvessels
cific immune response. Therefore, the T-cell of mice treated with either TNF- or bacterial endo-
response is likely to be antigen-nonspecific. Alter- toxin (Piccio et al., 2002), and in mice receiving CD4+
natively, lymphocytes may initiate and/or propa- and CD8+ T-lymphocytes obtained from patients with
gate the inflammatory response that occurs within multiple sclerosis (MS) (Battistini et al., 2003). These
the brain following stroke by releasing mediators studies have implicated a role for PSGL-1, a major
that increase the expression of endothelial CAMs counter-receptor for P-selectin, VCAM-1, and
and/or activate resident macrophages or other pop- CD11/CD18 in the lymphocyte recruitment process.
ulations of leukocytes. This possibility is supported Whether the same adhesion molecules mediate the
by the general view that lymphocytes enter the brain recruitment of lymphocytes into the postischemic cere-
in very small numbers early following reperfusion, bral microvasculature remains unclear.

NeuroMolecular Medicine Volume 7, 2005


234 Arumugam et al.

The 2-integrins expressed on T-cells allow these in postischemic brain tissue contribute to the recruit-
circulating cells to establish strong adhesive bonds ment of inflammatory cells that mediate some of
with vascular endothelium and enable leukocytes the I/R injury (Elneihoum et al., 1996). Further sup-
to remain stationary on the vessel wall. The 2-inte- port for a modulating role of TNF- in ischemic
grins and ICAM-1 are known to mediate the firm brain injury is provided by reports describing an
adhesion of leukocytes, including CD4+ and CD8+ exacerbation of MCAO-induced brain injury when
T-lymphocytes, in inflamed venules. There are sev- TNF- is administered by intra-cerebroventricular
eral lines of evidence that implicate the 2-integrins injection 24 h prior to the ischemic insult (Barone
in ischemic stroke. Expression of CD11a/CD18 on et al., 1997), and the reversal of this injury by intra-
circulating leukocytes is increased in patients with ventricular injection of an anti-TNF- monoclonal
ischemic stroke (Kim et al., 1995). Furthermore, pre- antibody (MAb). Treatment with the TNF- block-
treatment of mice with blocking antibodies directed ing MAb or soluble TNF-R1 has also been shown to
against CD11a, CD18, or CD11/CD18 reduce infarct reduce tissue damage and improve outcome in
size, edema formation, and neutrophil accumula- animal models of ischemic stroke (Barone et al., 1997;
tion after cerebral I/R (Matsuo et al., 1994; Yenari Nawashiro et al., 1997). Furthermore, selective inhi-
et al., 1998). Similarly, mice that are genetically defi- bition of TNF--converting enzyme, an enzyme that
cient in either CD11a, CD11b, CD18, or ICAM-1 are releases TNF- from its inactive cell-bound pre-
protected against ischemic brain injury (Prestigia- cursor, effectively suppresses the ischemia-induced
como et al., 1999; Soriano et al., 1999; Arumugam elevation in brain tissue TNF- levels and reduces
et al., 2004), suggesting that the leukocyte recruit- the infarct size and neurological deficit induced by
ment mediated by these adhesion molecules is crit- focal cerebral ischemia in the rat (Wang et al., 2004).
ical for the subsequent tissue injury. It remains However, the actions of TNF- in I/R injury are
unclear, however, whether the contribution of the complex, as this cytokine can also act directly on
2-integrins in ischemic stroke is entirely related to neurons to promote their survival through a nuclear
modulating the recruitment of neutrophils or factor (NF)-B-mediated mechanism (Gary et al.,
whether these adhesion molecules are linked to the 1998). Brain levels of IL-1 are also increased after
involvement of CD4+ and CD8+ T-lymphocytes in ischemia and may exacerbate ischemic brain injury
this injury process. Our recent findings strongly (Liu et al., 1993). Intra-cerebroventricular adminis-
implicate both lymphocytes and the 2-integrins in tration of recombinant IL-1 receptor antagonist
mediating the recruitment of leukocytes and markedly reduces brain damage induced by transient
platelets in cerebral venules, tissue injury (infarct ischemia (Mulcahy et al., 2003), and a similar level
volume), and organ dysfunction (neurological score) of protection has been demonstrated in mutant mice
that are associated with ischemic stroke (Arumugam that are genetically deficient in the IL-1-converting
et al., 2004). enzyme (Schielke et al., 1998). Finally, there is also
Several inflammatory cytokines such as TNF- evidence implicating chemokines, such as interferon
and IL-1 as well as chemokines (IL-8, CXCR3) have (IFN)-inducible protein-10 (IP-10), IFN--induced
been implicated in the pathogenesis of ischemic monokine, and CXC chemokines in acute ischemic
stroke. Support for the involvement of these medi- brain injury (Wang et al., 2000). All of these
ators of inflammation is provided by evidence that chemokines are ligands for the CXCR3 receptor
the expression of mRNA or protein for these agents (whose production is stimulated by IFN-) that is
is increased in brain tissue after exposure to ischemia expressed on T-lymphocytes. A major consequence
(Liu et al., 1994; Kostulas et al., 1999; Wang et al., of CXCR3 receptor activation is the recruitment of
2000). The elevated expression of mRNA or protein T-lymphocytes. Acute brain ischemia is associated
occurs shortly after occlusion (within 13 h) and with an increased expression of CXCR3 and a con-
precedes the influx of leukocytes after reperfusion. comitant accumulation of leukocytes in the affected
Because cytokines such as TNF- and IL-1 are brain tissue.
known to either prime or activate cerebrovascular IFN-, considered a key regulator of immune
endothelial cells and thereby enhance the surface and inflammatory responses and absent from normal
expression of endothelial CAMs, it is widely brain parenchyma, may play a role in T-cell recruit-
assumed that the elevated cytokine levels detected ment in stroke. During inflammatory conditions

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 235

such as MS, IFN- is produced by infiltrating T-cells IL-12, IFN-, and TNF-, whereas TH2 cytokines
and natural killer cells. The role of IFN- in perma- IL-4, IL-5, IL-10, and IL-13 are thought to have down-
nent MCAO is controversial, with one group demon- regulatory properties. IFN- is thought to have a
strating an elevated IFN- mRNAand another study key role in the pathogenesis of MS, and in EAE, TH1
unable to corroborate this response. IFN- from a T-cells are also strongly associated with clinical dis-
non-T-cell source may promote T-cell interactions ease (Neumann et al., 2002; Segal, 2003). In human
with the vascular wall (e.g., via upregulation of stroke or in experimental models of stroke, the pos-
CD40 which leads to upregulation of endothelial sible pathogenic role of CD4+ TH1 cells has so far
adhesion molecules and lymphocyte recruitment; received little attention, despite the abundance of
Omari and Dorovini-Zis, 2003). Adherent lympho- CD4+ TH1 lymphocytes within the lesions.
cytes may in turn release IFN- that activates the Although the CNS is isolated from the immune
vascular endothelium to support direct platelet con- system by the BBB, activated CD4+ cells easily infil-
tact. This contention is supported by our recent find- trate the CNS following cerebral I/R (Hickey and
ings that the CD40CD40L dyad is an important Kimura, 1988; Schroeter et al., 1994). Consequently,
component of the early leukocyte recruitment in a microglia have the opportunity to retain and further
stroke model (Ishikawa et al., 2005). stimulate CD4+ cells already primed to differentiate
into TH1 cells producing proinflammatory cytokines
(IL-2, IFN-, TNF-) or into TH2 cells producing
Possible Mechanisms cytokines that support antibody-mediated responses
of T-Cell-Mediated Stroke Injury (IL-4, IL-5, IL-10, lL-13) (Mosmann and Sad, 1996).
However, it is has only recently been shown that
Cytotoxic T-cells (CD8+) recognize their antigen antigen-inexperienced, and thus unprimed, T-cells
in the context of major histocompatibility (MHC) are also able to permeate the BBB following brain
class I antigens on the surface of respective target inflammation. However, these T-cells do not express
cells. In normal human brain tissue, class I antigens other T-cell activation markers, such as CD40 ligand,
are constitutively expressed on all cerebral endothe- and thus cannot interact with microglia through
lial cells and on some microglia cells (Traugott, 1987). CD40, the receptor for CD40 ligand. A recent study
Under inflammatory conditions, these MHC mole- by Fee et al. (2003) using traumatic brain injury sug-
cules may appear on any cell type of the nervous gests that CD4+ T-cells contribute to the acute brain
system (Vass and Lassmann, 1990), as their expres- damage following trauma; however, as in the case
sion is regulated by the intensity of the proinflam- of stroke injury the precise mechanism involved
matory stimulus (e.g., IFN- and TNF-) (Vass and remains to be defined. One possible mechanism is
Lassmann, 1990; Neumann et al., 1995; 1997) and that CD4+ T-cells participate in inducing apoptosis
the integrity of the target cell. Thus, in principle, all of brain cells by producing inflammatory cytokines,
cells of the nervous system may become targets for such as IL-2, IFN-, and TNF-, or via FAS-depen-
cytotoxic T-lymphocytes. Cytotoxic T-cells may kill dent mechanisms (McLaurin et al., 1995; Aktas et al.,
their target cells either through their cytotoxic gran- 2005; Chaparro-Huerta et al., 2005). In addition,
ules, containing perforin and granzymes, or through unregulated access of CD4+ T-cells to the CNS fol-
TNF family receptor-mediated cytotoxicity, as in the lowing ischemic stroke can lead to the dysregula-
interaction between Fas-receptor with Fas-ligand. tion of CD4+ cells, which then results in inappropriate
For these reasons, direct CD8+ T-cell-mediated cyto- tissue damage. This dysregulation might be a con-
toxicity could play a major role in mediating inflam- sequence of the constant lack or temporary absence
matory tissue damage in the CNS following brain of inflammatory mediators and costimulatory
I/R. In experimental models, MHC class II-specific molecules readily available to CD4+ T-cells in lym-
CD4+ cells have been shown to be instrumental in phoid tissues. There is now abundant evidence from
the clearance of virus infection in the CNS. CD4+ several laboratories that immune pathology in non-
T-cells can be categorized into T-helper 1 and 2 (TH1 lymphoid tissues, for example the intestine, involves
and TH2) subsets, based on their profiles of cytokine the development of a dysregulated TH1 response in
expression. CD4+ TH1 cells, which mediate EAE, various cytokine-deficient mouse models (Groux
secrete proinflammatory cytokines, including IL-2, and Powrie, 1999). IFN- and TNF--secreting CD4+

NeuroMolecular Medicine Volume 7, 2005


236 Arumugam et al.

TH1 cells were shown to be involved in intestinal ulatory cells, which both suppress TH1 cells. It has
lesions (Bregenholt and Claesson, 1998). Our work been reported that elderly patients with a history
and another recent study using traumatic brain of stroke have significantly lower median IL-10
injury model (Fee et al., 2003) provide evidence for levels compared with elderly without stroke (van
the possibility of dysregulated access of activated Exel et al., 2002; Vila et al., 2003). Within the brain,
CD4+ T-cells to the CNS during traumatic injury or IL-10 may deactivate macrophage-like cells and
cerebral I/R. Activated T-cells may also contribute astrocytes and thus limit their involvement in a
to the pathogenesis of ischemic stroke by accumu- secondary inflammatory process. Furthermore, IL-10
lating within the microcirculation and impairing cap- limits the role of glutamate cytotoxicity by inacti-
illary perfusion and exacerbating the hypoxic stress. vation of NF-B (Bachis et al., 2001), a transcription
factor that modulates inflammation and key regu-
latory proteins in cerebral ischemia. IL-10 also
Neuroprotective Role for T-Cells prevents adhesion and extravasion of leukocytes by
Following Stroke targeting the interface between the BBB (Kubes and
Ward, 2000). Moreover, TGF-1 is expressed in brain
Our recent studies and the observations of others following ischemia (Buisson et al., 2003) and may
in studies of experimental stroke, EAE, and in an play a role as an anti-inflammatory cytokine with
animal model for MS, clearly point to a primary role neuroprotective properties. TGF-1 has been shown
for T-cells in the pathogenesis of brain disease. How- to protect cultured neurons from hypoxic (Prehn
ever, despite their pathogenic potential, a neuro- et al., 1993), excitotoxic (Prehn and Krieglstein,
protective role for T-cells after CNS injury has been 1994), apoptotic (Prehn et al., 1994), and toxic
suggested. In different experimental models of brain peptide-induced injury (Meucci and Miller, 1996).
injury, it has been shown that T-cells are capable of In vivo, administration of recombinant TGF-1 or
protecting nervous tissue from secondary injury gene delivery of TGF-1 potently protects animals
(Schwartz and Hauben, 2002). Autoimmune T-cells against brain injury mediated by ischemic (McNeill
specific for MBP could prevent neurons from the et al., 1994), excitotoxic (Ruocco et al., 1999), and
spread of neuronal death caused by primary injury transient global ischemic injury (Henrich-Noack et
to the spinal cord (Moalem et al., 1999). T-cells spe- al., 1996). Neuroprotection by TGF- 1 has been
cific for Copaxone 1, a compound used for MS ther- shown to involve the upregulation of antiapoptotic
apy, decreases secondary degeneration after optic Bcl-2 family proteins Bcl-2 and Bcl-xL (Kim et al.,
nerve crush (Kipnis et al., 2000). Passive and active 1998). These are target genes of the transcription
immunization with myelin peptides is also protec- factor NF-B and that neuroprotection elicited by
tive against neuronal death in spinal cord injury TGF-1 and TNF- requires NF-B activation (Matt-
(Hauben et al., 2000). In addition, it has been shown son et al., 1997; Zhu et al., 2004). However, it is also
that MBP-specific T-cells prevent cyst formation in important to note here that NF-B activation fol-
a spinal cord injury model (Butovsky et al., 2001). lowing brain ischemia may also be involved in the
These findings imply that T-cell responses against activation of proinflammatory genes. Other neuro-
specific self-antigens are not necessarily detrimen- protective mechanisms mediated by T-cells follow-
tal, and may have a neuroprotective role following ing stroke may be explained by anti-inflammatory
cerebral ischemia. cytokines and growth factors produced by TH2
Because cell death following stroke might take T-cells such as IL-4, IL-5, and IL-13. However, there
several days, the prime goal of neuroprotection is is no well-documented evidence for this.
to salvage the ischemic inflammation at the penum-
bra in which most of the apoptotic death occurs.
Anti-inflammatory responses of T-cells can be medi- Therapeutic Strategies to Target
ated by cytokines including IL-10 and transform- T-Cells in Ischemic Stroke
ing growth factor (TGF)-1 (Yoshimura et al., 2003;
OGarra et al., 2004; Grutz, 2005). IL-10 is preferen- Many therapeutic strategies for autoimmune dis-
tially produced by Tr1-type regulatory T-cells, and eases and neurodegenerative disorders are based
TGF-1 is preferentially produced by TH2-type reg- on induction of immune regulation with the aim of

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 237

altering or downregulating the phenotype of the humoral or cytotoxic immune response against the
T-cell-mediated response. According to the studies medication because of its murine origin or a non-
involving T-cells and ischemic stroke, clinical treat- specific complement activation induced by presen-
ment protocols targeting T-cells would have to be tation of Fc fragments (Furuya et al., 2001).
carefully designed to target both deleterious and Another way of targeting T-cell adhesion during
protective actions of T-cells and also on their nat- cerebral I/R is to administer molecules or antibod-
ural susceptibility to disease development. There- ies that target LFA-1. Our previous findings impli-
fore, regulation-based therapies targeting T-cells for cate a role for LFA-1 (CD11a/CD18) and Mac-1
stroke-related pathologies should be designed and (CD11b/CD18) in the recruitment of leukocytes in
administered with caution. There are many strate- cerebral venules following ischemic stroke (Aru-
gies to target T-cell-mediated pathogenesis in stroke mugam et al., 2004). Efalizumab (Raptiva, anti-
such as T-cell receptor (TCR) antagonism, admin- CD11a) is a humanized form of a murine antibody
istration of antiadhesion molecules, and anticy- directed against CD11a, the -subunit of LFA-1. In
tokine treatment. The TCR recognizes an antigen as vitro studies demonstrated that by binding to
a short peptide bound to a major MHC molecule on CD11a, Efalizumab inhibits multiple key pathogenic
the surface of APCs. As pointed out earlier, inter- steps: T-cell activation (Werther et al., 1996), cuta-
action between the TCR and MHCpeptide com- neous T-cell trafficking (Gottlieb et al., 2000), and
plex leads to activation of T-cells. T-cell responses T-cell adhesion. These findings suggest that Efal-
to MHC molecules during cerebral ischemia or izumab exerts its clinical activity via several distinct
during reperfusion and its deleterious effects can mechanisms. A small molecule antagonist of LFA-1
be inhibited by introducing subtle changes in the (BIRT 377) that inhibits LFA-1ICAM-1 molecular
antigenic peptide amino acid content. Certain interactions has also been developed and tested
analogs of an antigenic peptide containing single against different disease conditions that involve T-
amino acid substitutions in the TCR contact residues cells (Kapadia et al., 2001). However, neither Efal-
fail to stimulate T-cell responses on their own, but izumab nor BIRT 377 has been used in preclinical
are able to diminish, or even inhibit, T-cell activa- or clinical studies of stroke.
tion induced by the original peptide. The inhibitory Clearly, proinflammatory cytokines such as
actions of TCR antagonists have been shown not TNF-, IFN-, IL-1, and IL-6 produced by T-cells
only in vitro but also in vivo (Kuchroo et al., 1994; are involved in the pathogenesis of stroke and some
Williams et al., 1998; Toda et al., 2000). However, TH2-specific anti-inflammatory cytokines such as IL-
the effect of TCR antagonists on the primary 10 and IL-4 might exert beneficial effects as explained
responses of T-cells following cerebral ischemia has in the previous section. Targeting T-cell-specific
not yet been determined. proinflammatory cytokines without affecting ben-
The crucial role of adhesion molecules in the eficial cytokines might be another way of treating
pathogenesis of stroke makes them interesting tar- stroke. However, it should be noted here that most
gets for drug development. Studies with antiadhe- of these proinflammatory cytokines produced
sion strategies have been undertaken against many during stroke are produced by macrophages and
inflammatory diseases in which T-cells involve- glial cells rather than T-cells. Thus, targeting inflam-
ment is crucial in the disease development. However, matory cytokines may be an effective option for the
there have been a remarkable number of therapeu- therapy of stroke. In this regard, TNF-, IL-1, IL-6,
tic failures involved with clinically introduced anti- IL-18, and IFN- may represent interesting targets
adhesion therapies such as anti-ICAM-1 antibody for the treatment of stroke.
(Enlimomab Acute Stroke Trial Investigators, 2001). In conclusion, the brain microvasculature after a
Treatment with a murine anti-ICAM-1 antibody was stroke assumes an inflammatory phenotype that is
investigated in patients with acute ischemic stroke characterized by leukocyteendothelial cell adhe-
in the Enlimomab acute stroke trial but, unfortu- sion, leukocyte capillary plugging, endothelial bar-
nately, the case fatality rate was significantly higher rier dysfunction, activation of resident macrophages,
in the Enlimomab patient group than in the placebo and an enhanced generation of oxidants and
group. Adverseeffects of Enlimomab may have been inflammatory mediators. T-lymphocytes also accu-
caused by immunological factors, such as a specific mulate in the postischemic brain and contribute to

NeuroMolecular Medicine Volume 7, 2005


238 Arumugam et al.

tissue damage by several mechanisms. Cytotoxic T- haematopoiesis in severe combined immunodefi-


cells may play deleterious roles either through their cient (scid) mice with inflammatory bowel disease
cytotoxic granules, containing perforin and (IBD). Clin. Exp. Immunol. 111, 166172.
granzymes, or through TNF family receptor-medi- Buisson A., Lesne S., Docagne F., et al. (2003) Trans-
ated cytotoxicity. CD4+ cells also infiltrate the CNS forming growth factor-beta and ischemic brain
following stroke and contribute to the acute brain injury. Cell Mol. Neurobiol. 23, 539550.
injury by producing proinflammatory cytokines. Burne-Taney M. J., Kofler J., Yokota N., Weisfeldt M.,
On the other hand anti-inflammatory responses of Traystman R. J., and Rabb H. (2003) Acute renal
T-cells can be mediated by cytokines including IL- failure after whole body ischemia is characterized
10 and TGF-1 preferentially produced by TH2-type by inflammation and T cell-mediated injury. Am.
J. Physiol. Renal Physiol. 285, F87F94.
regulatory cells. Clinical treatments targeting T-cells
Burne-Taney M. J., Yokota-Ikeda N., and Rabb H. (2005)
may have beneficial effects against stroke injury, but
Effects of combined T- and B-cell deficiency on
must be designed to differentially target the dele- murine ischemia reperfusion injury. Am. J. Trans-
terious and protective actions of T-cells. plant. 5, 11861193.
Butovsky O., Hauben E., and Schwartz M. (2001) Mor-
phological aspects of spinal cord autoimmune neu-
References roprotection: colocalization of T cells with B72
(CD86) and prevention of cyst formation. FASEB J.
Aktas O., Smorodchenko A., Brocke S., et al. (2005) 15, 10651067.
Neuronal damage in autoimmune neuroinflam- Campanella M., Sciorati C., Tarozzo G., and Beltramo
mation mediated by the death ligand TRAIL. M. (2002) Flow cytometric analysis of inflamma-
Neuron 46, 421432. tory cells in ischemic rat brain. Stroke 33, 586592.
Arumugam T. V., Salter J. W., Chidlow, J. H., Ballan- Carden D. L. and Granger D. N. (2000) Pathophysiol-
tyne C. M., Kevil C. G., and Granger D. N. (2004) ogy of ischaemia-reperfusion injury. J. Pathol. 190,
Contributions of LFA-1 and Mac-1 to brain injury 255266.
and microvascular dysfunction induced by tran- Chaparro-Huerta V., Rivera-Cervantes M. C., Flores-
sient middle cerebral artery occlusion. Am. J. Soto M. E., Gomez-Pinedo U., and Beas-Zarate C.
Physiol. Heart. Circ. Physiol. 287, H2555H2560. (2005) Proinflammatory cytokines and apoptosis
Bachis A., Colangelo A. M., Vicini S., Doe P. P., De following glutamate-induced excitotoxicity medi-
Bernardi M. A., Brooker G., and Mocchetti I. (2001) ated by p38 MAPK in the hippocampus of neona-
Interleukin-10 prevents glutamate-mediated cere- tal rats. J. Neuroimmunol. 165, 5362.
bellar granule cell death by blocking caspase-3- Chavarria A. and Alcocer-Varela J. (2004) Is damage
like activity. J. Neurosci. 21, 31043112. in central nervous system due to inflammation?
Barone F. C., Arvin B., White R. F., et al. (1997) Tumor Autoimmun. Rev. 3, 251260.
necrosis factor-alpha. Amediator of focal ischemic DeGraba T. J. (1998) The role of inflammation after
brain injury. Stroke 28, 12331244. acute stroke: utility of pursuing anti-adhesion mol-
Battistini L., Piccio L., Rossi B., et al. (2003) CD8+ T ecule therapy. Neurology 51, S62S68.
cells from patients with acute multiple sclerosis Dirnagl U., Simon R. P., and Hallenbeck J. M. (2003)
display selective increase of adhesiveness in brain Ischemic tolerance and endogenous neuroprotec-
venules: a critical role for P-selectin glycoprotein tion. Trends Neurosci. 26, 248254.
ligand-1. Blood 101, 47754782. Elneihoum A. M., Falke P., Axelsson L., Lundberg E.,
Bochelen D., Rudin M., and Sauter A. (1999) Calcineurin Lindgarde F., and Ohlsson K. (1996) Leukocyte
inhibitors FK506 and SDZ ASM 981 alleviate the activation detected by increased plasma levels
outcome of focal cerebral ischemic/reperfusion of inflammatory mediators in patients with
injury. J. Pharmacol. Exp. Ther. 288, 653659. ischemic cerebrovascular diseases. Stroke 27,
Brecht S., Schwarze K., Waetzig V., et al. (2003) Changes 17341738.
in peptidyl-prolyl cis/trans isomerase activity and Enlimomab Acute Stroke Trial Investigators. (2001)
FK506 binding protein expression following neu- Use of anti-ICAM-1 therapy in ischemic stroke:
roprotection by FK506 in the ischemic rat brain. results of the Enlimomab Acute Stroke Trial. Neu-
Neuroscience 120, 10371048. rology 57, 14281434.
Bregenholt S. and Claesson M. H. (1998) Splenic T helper Fee D., Crumbaugh A., Jacques T., et al. (2003) Acti-
cell type 1 cytokine profile and extramedullary vated/effector CD4+ T cells exacerbate acute

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 239

damage in the central nervous system following Hickey W. F. and Kimura H. (1988) Perivascular
traumatic injury. J. Neuroimmunol. 136, 5466. microglial cells of the CNS are bone marrow-
Flad H. D., Harter L., Petersen F., et al. (1997) Regula- derived and present antigen in vivo. Science 239,
tion of neutrophil activation by proteolytic pro- 290292.
cessing of platelet-derived alpha-chemokines. Adv. Hogg N., Smith A., McDowall A., Giles K., Stanley P.,
Exp. Med. Biol. 421, 223230. Laschinger M., and Henderson R. (2004) How T
Frijns C. J. and Kappelle L. J. (2002) Inflammatory cell cells use LFA-1 to attach and migrate. Immunol.
adhesion molecules in ischemic cerebrovascular Lett. 92, 5154.
disease. Stroke 33, 21152122. Ishikawa M., Cooper D., Arumugam T. V., Zhang J. H.,
Furuya K., Takeda H., Azhar S., et al. (2001) Exami- Nanda A., and Granger D. N. (2004a) Platelet-
nation of several potential mechanisms for the neg- leukocyte-endothelial cell interactions after
ative outcome in a clinical stroke trial of middle cerebral artery occlusion and reperfusion.
enlimomab, a murine anti-human intercellular J. Cereb. Blood Flow Metab. 24, 907915.
adhesion molecule-1 antibody: a bedside-to-bench Ishikawa M., Cooper D., Russell J., Salter J. W., Zhang
study. Stroke 32, 26652674. J. H., Nanda A., and Granger D. N. (2003) Molec-
Garlichs C. D., Kozina S., Fateh-Moghadam S., et al. ular determinants of the prothrombogenic and
(2003) Upregulation of CD40-CD40 ligand (CD154) inflammatory phenotype assumed by the postis-
in patients with acute cerebral ischemia. Stroke 34, chemic cerebral microcirculation. Stroke 34,
14121418. 17771782.
Gary D. S., Bruce-Keller A. J., Kindy M. S., and Matt- Ishikawa M., Vowinkel T., Stokes K. Y., Arumugam T.
son M. P. (1998) Ischemic and excitotoxic brain V., Yilmaz G., Nanda A., and Granger D. N. (2005)
injury is enhanced in mice lacking the p55 tumor CD40/CD40 ligand signaling in mouse cerebral
necrosis factor receptor. J. Cereb. Blood Flow Metab. microvasculature after focal ischemia/reperfu-
18, 12831287. sion. Circulation 111, 16901696.
Gottlieb A., Krueger J. G., Bright R., et al. (2000) Effects Ishikawa M., Zhang J. H., Nanda A., and Granger D.
of administration of a single dose of a humanized N. (2004b) Inflammatory responses to ischemia
monoclonal antibody to CD11a on the immuno- and reperfusion in the cerebral microcirculation.
biology and clinical activity of psoriasis. J. Am. Front. Biosci. 9, 13391347.
Acad. Dermatol. 42, 428435. Jander S., Kraemer M., Schroeter M., Witte O. W., and
Groux H. and Powrie F. (1999) Regulatory T cells and Stoll G. (1995) Lymphocytic infiltration and expres-
inflammatory bowel disease. Immunol. Today 20, sion of intercellular adhesion molecule-1 in pho-
442445. tochemically induced ischemia of the rat cortex.
Grutz G. (2005) New insights into the molecular mech- J. Cereb. Blood Flow Metab. 15, 4251.
anism of interleukin-10-mediated immunosup- Kapadia S. R., Spero D. M., and Eriksson M. (2001) An
pression. J. Leukoc. Biol. 77, 315. improved synthesis of chiral alpha-(4-bromoben-
Guo Z., Iyun T., Fu W., Zhang P., and Mattson M. P. zyl)alanine ethyl ester and its application to the
(2004) Bone marrow transplantation reveals roles synthesis of LFA-1 antagonist BIRT-377. J. Org.
for brain macrophage/microglia TNF signaling Chem. 66, 19031905.
and nitric oxide production in excitotoxic neuronal Kim E. S., Kim R. S., Ren R. F., Hawver D. B., and Flan-
death. Neuromol. Med. 5, 219234. ders K. C. (1998) Transforming growth factor-beta
Han H. S. and Yenari M. A. (2003) Cellular targets of inhibits apoptosis induced by beta-amyloid pep-
brain inflammation in stroke. Curr. Opin. Investig. tide fragment 2535 in cultured neuronal cells.
Drugs 4, 522529. Brain Res. Mol. Brain Res. 62, 122130.
Hauben E., Butovsky O., Nevo U., et al. (2000) Passive Kim J. S., Chopp M., Chen H., Levine S. R., Carey J. L.,
or active immunization with myelin basic protein and Welch K. M. (1995) Adhesive glycoproteins
promotes recovery from spinal cord contusion. J. CD11a and CD18 are upregulated in the leuko-
Neurosci. 20, 64216430. cytes from patients with ischemic stroke and tran-
Henrich-Noack P., Prehn J. H., and Krieglstein J. sient ischemic attacks. J. Neurol. Sci. 128, 4550.
(1996) TGF-beta 1 protects hippocampal neurons Kipnis J., Yoles E., Porat Z., et al. (2000) T cell immu-
against degeneration caused by transient global nity to copolymer 1 confers neuroprotection on
ischemia. Dose-response relationship and poten- the damaged optic nerve: possible therapy for optic
tial neuroprotective mechanisms. Stroke 27, neuropathies. Proc. Natl. Acad. Sci. USA 97,
16091614. 74467451.

NeuroMolecular Medicine Volume 7, 2005


240 Arumugam et al.

Kostulas N., Pelidou S. H., Kivisakk P., Kostulas V., Mosmann T. R. and Sad S. (1996) The expanding uni-
and Link H. (1999) Increased IL-1beta, IL-8, and verse of T-cell subsets: Th1, TH2 and more.
IL-17 mRNA expression in blood mononuclear Immunol. Today 17, 138146.
cells observed in a prospective ischemic stroke Mulcahy N. J., Ross J., Rothwell N. J., and Loddick S.
study. Stroke 30, 21742179. A. (2003) Delayed administration of interleukin-
Kubes P. and Ward P. A. (2000) Leukocyte recruitment 1 receptor antagonist protects against transient
and the acute inflammatory response. Brain Pathol. cerebral ischaemia in the rat. Br. J. Pharmacol. 140,
10, 127135. 471476.
Kuchroo V. K., Greer J. M., Kaul D., et al. (1994) Asingle Nawashiro H., Martin D., and Hallenbeck J. M. (1997)
TCR antagonist peptide inhibits experimental Inhibition of tumor necrosis factor and ameliora-
allergic encephalomyelitis mediated by a diverse tion of brain infarction in mice. J. Cereb. Blood Flow
T cell repertoire. J. Immunol. 153, 33263336. Metab. 17, 229232.
Le Moine O., Louis H., Demols A., et al. (2000) Cold Neumann H., Cavalie A., Jenne D. E., and Wekerle H.
liver ischemia-reperfusion injury critically (1995) Induction of MHC class I genes in neurons.
depends on liver T cells and is improved by donor Science 269, 549552.
pretreatment with interleukin 10 in mice. Hepa- Neumann H., Medana I. M., Bauer J., and Lassmann
tology 31, 12661274. H. (2002) Cytotoxic T lymphocytes in autoimmune
Liu T., Clark R. K., McDonnell P. C., Young P. R., White and degenerative CNS diseases. Trends Neurosci.
R. F., Barone F. C., and Feuerstein G. Z. (1994) 25, 313319.
Tumor necrosis factor-alpha expression in ischemic Neumann H., Schmidt H., Cavalie A., Jenne D., and
neurons. Stroke 25, 14811488. Wekerle H. (1997) Major histocompatibility complex
Liu T., McDonnell P. C., Young P. R., et al. (1993) Inter- (MHC) class I gene expression in single neurons
leukin-1 beta mRNA expression in ischemic rat of the central nervous system: differential regula-
cortex. Stroke 24, 17461750. tion by interferon (IFN)-gamma and tumor necrosis
Matsuo Y., Onodera H., Shiga Y., et al. (1994) Role of factor (TNF)-alpha. J. Exp. Med. 185, 305316.
cell adhesion molecules in brain injury after tran- OGarra A., Vieira P. L., Vieira P., and Goldfeld A. E.
sient middle cerebral artery occlusion in the rat. (2004) IL-10-producing and naturally occurring
Brain Res. 656, 344352. CD4+ Tregs: limiting collateral damage. J. Clin.
Mattson M. P., Goodman Y., Luo H., Fu W., and Invest. 114, 13721378.
Furukawa K. (1997) Activation of NF-kappaB pro- Omari K. M. and Dorovini-Zis K. (2003) CD40
tects hippocampal neurons against oxidative expressed by human brain endothelial cells regu-
stress-induced apoptosis: evidence for induction lates CD4+ T cell adhesion to endothelium. J. Neu-
of manganese superoxide dismutase and sup- roimmunol. 134, 166178.
pression of peroxynitrite production and protein Phillis J. W., Diaz F. G., ORegan M. H., and Pilitsis J.
tyrosine nitration. J. Neurosci. Res. 49, 681697. G. (2002) Effects of immunosuppressants, calcineurin
McLaurin J., DSouza S., Stewart J., et al. (1995) Effect inhibition, and blockade of endoplasmic reticu-
of tumor necrosis factor alpha and beta on human lum calcium channels on free fatty acid efflux from
oligodendrocytes and neurons in culture. Int. J. the ischemic/reperfused rat cerebral cortex. Brain
Dev. Neurosci. 13, 369381. Res. 957, 1224.
McNeill H., Williams C., Guan J., et al. (1994) Neu- Piccio L., Rossi B., Scarpini E., et al. (2002) Molecular
ronal rescue with transforming growth factor-beta mechanisms involved in lymphocyte recruitment
1 after hypoxic-ischaemic brain injury. Neuroreport in inflamed brain microvessels: critical roles for
5, 901904. P-selectin glycoprotein ligand-1 and het-
Meucci O. and Miller R. J. (1996) gp120-induced neu- erotrimeric G(i)-linked receptors. J. Immunol. 168,
rotoxicity in hippocampal pyramidal neuron cul- 19401949.
tures: protective action of TGF-beta1. J. Neurosci. Prehn J. H., Bindokas V. P., Marcuccilli C. J., Krajew-
16, 40804088. ski S., Reed J. C., and Miller R. J. (1994) Regula-
Moalem G., Leibowitz-Amit R., Yoles E., Mor F., Cohen tion of neuronal Bcl2 protein expression and
I. R., and Schwartz M. (1999) Autoimmune T cells calcium homeostasis by transforming growth
protect neurons from secondary degeneration after factor type beta confers wide-ranging protection
central nervous system axotomy. Nat. Med. 5, on rat hippocampal neurons. Proc. Natl. Acad. Sci.
4955. USA 91, 12,59912,603.

NeuroMolecular Medicine Volume 7, 2005


Stroke and T-Cells 241

Prehn J. H. and Krieglstein J. (1994) Opposing effects Stoll G., Muller S., Schmidt B., van der Meide P., Jung
of transforming growth factor-beta 1 on glutamate S., Toyka K. V., and Hartung H. P. (1993) Local-
neurotoxicity. Neuroscience 60, 710. ization of interferon-gamma and Ia-antigen in
Prehn J. H., Peruche B., Unsicker K., and Krieglstein T cell line-mediated experimental autoimmune
J. (1993) Isoform-specific effects of transform- encephalomyelitis. Am. J. Pathol. 142, 1866
ing growth factors-beta on degeneration of pri- 1875.
mary neuronal cultures induced by cytotoxic Tailor A. and Granger D. N. (2000) Role of adhesion
hypoxia or glutamate. J. Neurochem. 60, molecules in vascular regulation and damage.
16651672. Curr. Hypertens Rep. 2, 7883.
Prestigiacomo C. J., Kim S. C., Connolly E. S., Jr., Liao Toda M., Totsuka M., Furukawa S., et al. (2000) Down-
H., Yan S. F., and Pinsky D. J. (1999) CD18-medi- regulation of antigen-specific antibody produc-
ated neutrophil recruitment contributes to the tion by TCR antagonist peptides in vivo. Eur. J.
pathogenesis of reperfused but not nonreperfused Immunol. 30, 403414.
stroke. Stroke 30, 11101117. Traugott U. (1987) Multiple sclerosis: relevance of class
Price C. J., Warburton E. A., and Menon D. K. (2003) I and class II MHC-expressing cells to lesion devel-
Human cellular inflammation in the pathology of opment. J. Neuroimmunol. 16, 283302.
acute cerebral ischaemia. J. Neurol. Neurosurg. Psy- van Exel E., Gussekloo J., de Craen A. J., Bootsma-van
chiatry 74, 14761484. der Wiel A., Frolich M., and Westendorp R. G.
Ruocco A., Nicole O., Docagne F., et al. (1999) A trans- (2002) Inflammation and stroke: the Leiden 85-
forming growth factor-beta antagonist unmasks Plus Study . Stroke 33, 11351138.
the neuroprotective role of this endogenous Vandendries E. R., Furie B. C., and Furie B. (2004) Role
cytokine in excitotoxic and ischemic brain injury. of P-selectin and PSGL-1 in coagulation and throm-
J. Cereb. Blood Flow Metab. 19, 13451353. bosis. Thromb. Haemost. 92, 459466.
Schielke G. P., Yang G. Y., Shivers B. D., and Betz A. L. Vass K. and Lassmann H. (1990) Intrathecal applica-
(1998) Reduced ischemic brain injury in inter- tion of interferon gamma. Progressive appearance
leukin-1 beta converting enzyme-deficient mice. of MHC antigens within the rat nervous system.
J. Cereb. Blood Flow Metab. 18, 180185. Am. J. Pathol. 137, 789800.
Schroeter M., Jander S., Witte O. W., and Stoll G. (1994) Vemuganti R., Dempsey R. J., and Bowen K. K. (2004)
Local immune responses in the rat cerebral cortex Inhibition of intercellular adhesion molecule-1
after middle cerebral artery occlusion. J. Neuroim- protein expression by antisense oligonucleotides
munol. 55, 195203. is neuroprotective after transient middle cerebral
Schwartz M. and Hauben E. (2002) T cell-based ther- artery occlusion in rat. Stroke 35, 179184.
apeutic vaccination for spinal cord injury. Prog. Vila N., Castillo J., Davalos A., Esteve A., Planas A. M.,
Brain Res. 137, 401406. and Chamorro A. (2003) Levels of anti-inflamma-
Segal B. M. (2003) Experimental autoimmune tory cytokines and neurological worsening in acute
encephalomyelitis: cytokines, effector T cells, and ischemic stroke. Stroke 34, 671675.
antigen-presenting cells in a prototypical Th1- Wang X., Feuerstein G. Z., Xu L., et al. (2004) Inhibi-
mediated autoimmune disease. Curr. Allergy tion of tumor necrosis factor-alpha-converting
Asthma Rep. 3, 8693. enzyme by a selective antagonist protects brain
Shigematsu T., Wolf R. E., and Granger D. N. (2002) T- from focal ischemic injury in rats. Mol. Pharmacol.
lymphocytes modulate the microvascular and 65, 890896.
inflammatory responses to intestinal ischemia- Wang X., Li X., Schmidt D. B., et al. (2000) Identifica-
reperfusion. Microcirculation 9, 99109. tion and molecular characterization of rat CXCR3:
Soriano S. G., Coxon A., Wang Y. F., et al. (1999) Mice receptor expression and interferon-inducible pro-
deficient in Mac-1 (CD11b/CD18) are less suscep- tein-10 binding are increased in focal stroke. Mol.
tible to cerebral ischemia/reperfusion injury. Stroke Pharmacol. 57, 11901198.
30, 134139. Ward P. A., Warren J. S., and Johnson K. J. (1988) Oxygen
Stevens S. L., Bao J., Hollis J., Lessov N. S., Clark W. radicals, inflammation, and tissue injury. Free
M., and Stenzel-Poore M. P. (2002) The use of flow Radic. Biol. Med. 5, 403408.
cytometry to evaluate temporal changes in inflam- Werther W. A., Gonzalez T. N., OConnor S. J., et al. (1996)
matory cells following focal cerebral ischemia in Humanization of an anti-lymphocyte function-asso-
mice. Brain Res. 932, 110119. ciated antigen (LFA)-1 monoclonal antibody and

NeuroMolecular Medicine Volume 7, 2005


242 Arumugam et al.

reengineering of the humanized antibody for bind- influences inflammation. Curr. Opin. Immunol. 15,
ing to rhesus LFA-1. J. Immunol. 157, 49864995. 704708.
Whatley R. E., Zimmerman G. A., McIntyre T. M., and Ysebaert D. K., De Greef K. E., De Beuf A., et al. (2004)
Prescott S. M. (1990) Lipid metabolism and signal trans- T cells as mediators in renal ischemia/reperfusion
duction in endothelial cells. Prog. Lipid Res. 29, 4563. injury. Kidney Int. 66, 491496.
Williams C. B., Vidal K., Donermeyer D., Peterson D. Zhu Y., Culmsee C., Klumpp S., and Krieglstein J. (2004)
A., White J. M., and Allen P. M. (1998) In vivo Neuroprotection by transforming growth factor-
expression of a TCR antagonist: T cells escape beta1 involves activation of nuclear factor-kappaB
central tolerance but are antagonized in the periph- through phosphatidylinositol-3-OH kinase/Akt
ery. J. Immunol. 161, 128137. and mitogen-activated protein kinase-extracellu-
Yenari M. A., Kunis D., Sun G. H., et al. (1998) Hu23F2G, lar-signal regulated kinase1,2 signaling pathways.
an antibody recognizing the leukocyte CD11/CD18 Neuroscience 123, 897906.
integrin, reduces injury in a rabbit model of tran- Zwacka R. M., Zhang Y., Halldorson J., Schlossberg
sient focal cerebral ischemia. Exp. Neurol. 153, H., Dudus L., and Engelhardt J. F. (1997) CD4(+)
223233. T-lymphocytes mediate ischemia/reperfusion-
Yoshimura A., Mori H., Ohishi M., Aki D., and Hanada induced inflammatory responses in mouse liver.
T. (2003) Negative regulation of cytokine signaling J. Clin. Invest. 100, 279289.

NeuroMolecular Medicine Volume 7, 2005

S-ar putea să vă placă și