Vous êtes sur la page 1sur 7

Stroke-Induced Immunodepression

Experimental Evidence and Clinical Relevance


Ulrich Dirnagl, MD; Juliane Klehmet, MD; Johann S. Braun, MD; Hendrik Harms, MD;
Christian Meisel, MD; Tjalf Ziemssen, MD; Konstantin Prass, MD; Andreas Meisel, MD

Abstract—Stroke affects the normally well-balanced interplay of the 2 supersystems: the nervous and the immune system.
Recent research elucidated some of the involved signals and mechanisms and, importantly, was able to demonstrate that
brain-immune interactions are highly relevant for functional outcome after stroke. Immunodepression after stroke
increases the susceptibility to infection, the most relevant complication in stroke patients. However, immunodepression
after stroke may also have beneficial effects, for example, by suppressing autoaggressive responses during lesion-
induced exposure of central nervous system-specific antigens to the immune system. Thus, before immunomodulatory
therapy can be applied to stroke patients, we need to understand better the interaction of brain and immune system after
focal cerebral ischemia. Until then, anticipating an important consequence of stroke-induced immunodepression,
bacterial infection, preventive antibiotic strategies have been proposed. In mouse experiments, preventive antibiotic
treatment dramatically improves mortality and outcome. Results of clinical studies on this issue are contradictory at
present, and larger trials are needed to settle the question whether (and which) stroke patients should be preventively
treated. Nevertheless, clinical evidence is emerging demonstrating that stroke-induced immunodepression in humans not
only exists, but has very similar features to those characterized in rodent experiments. (Stroke. 2007;38[part 2]:770-
773.)
Key Words: brain infarction brain ischemia experime tal focal ischemia immu ology infectious disease
inflammation treatment

R ecently, major advances have been made in our under-


standing of the roles of both cellular and humoral
inflammation in cerebral ischemia. In particular, it is emerg-
neurons) transform to an inflammatory phenotype. Most of
the available literature links stroke induced inflammation to
the progression of damage. Thus, inhibition of inflammation
ing that innate and adaptive immunity play an important role would seem to be the most straightforward therapeutic
for the outcome after focal cerebral ischemia (stroke). Stroke strategy. However, inflammation is a key mechanism of any
has dramatic consequences for the normally well-balanced tissue to cope not only with the invasion of pathogens but also
interplay of the two supersystems, the nervous and the with tissue destruction. Inflammation plays an important part
immune systems: brain inflammation and immunodepression. in the clearing of damaged tissue, and in the ensuing pro-
We are only beginning to understand that the consequences of cesses of angiogenesis, tissue remodeling, and regeneration.
brain-immune interactions after stroke are neither “good” nor This is probably best studied in wound healing, which is
“bad,” an insight that might imply that targeted therapeutic severely compromised if inflammation is inhibited.1,2 The
intervention (“anti-inflammation”; “immunomodulation,” potential benefits of inflammation after stroke have received
etc) may prove to be less straightforward as initially hoped relatively little attention so far, but indirect evidence suggests
for. Here we briefly review stroke-induced inflammation, and that certain kinds of inflammatory reactions are neuroprotec-
focus on stroke-induced immunodepression, a recently de- tive and neuroregenerative.3,4 In brief, macrophages and
scribed phenomenon that may be the key factor underlying microglia produce a host of trophic cytokines when activated,
susceptibility to infection after stroke. and macrophages or T-cells exposed to certain central ner-
vous system (CNS)-specific antigens ex vivo partake in tissue
Stroke Induces Local Inflammation In Situ repair and recovery after nerve transsection and spinal cord
After stroke, leukocytes home toward the lesion, and brain injury. However, these benefits may be at least partially offset
parenchymal cells (microglia, astrocytes, endothelia, even not only by “bystander toxicity” of inflammation but also by

Received May 17, 2006; final revision received September 4, 2006; accepted September 13, 2006.
From Department of Neurology (U.D., J.K., J.S.B., H.H., K.P., A.M.), Department of Experimental Neurology, Center for Stroke Research and
Department of Medical Immunology (C.M.), Charité Universitätsmedizin, Berlin, Germany; Department of Neurology (T.Z.), University of Dresden,
Germany.
Correspondence to Prof Dr Ulrich Dirnagl, Department of Experimental Neurology, Charite Universitätsmedizin Berlin, Charitéplatz 1, 10098 Berlin,
Germany. E-mail ulrich.dirnagl@charite.de
© 2007 American Heart Association, Inc.
Stroke is available at http://www.strokeaha.org DOI: 10.1161/01.STR.0000251441.89665.bc

Downloaded from http://stroke.ahaj7o7u0rnals.org/ by guest on May 24, 2015


Dirnagl et al Stroke-Induced Immunodepression 771
Adoptive transfer of T and natural killer cells from wild-type
mice, but not from interferon (IFN) -deficient mice, or

Figure 1. Experimental stroke induces immunodepression via


the hypothalamic pituitary axis (HPA) and the sympathetic ner-
vous system (SNS). SNS plays a key role because -receptor
blockade exerts comprehensive effects on stroke induced alter-
ations of the immune system and outcome (box). Blockade of
the HPA axis with the glucocorticoid receptor antagonist RU486
only improves lymphopenia and monocyte deactivation.9

scar formation, which in peripheral tissues is key to wound


closure, but in the brain is a major impediment of regenera-
tion and plasticity.5 A recent example for the dual nature and
complexities of ischemia-induced inflammation has been the
uncovering of a regenerative role for matrix metalloprotein
activation,6 enzymes that were previously almost exclusively
linked to blood– brain barrier disruption and lesion expansion.

Stroke Induces Systemic Immunodepression


Infections are a leading cause of death in patients with stroke.
Eighty-five percent of all stroke patients have relevant com-
plications; of which, infection is the most frequent (23% to
65%). Infection also is the most relevant complication during
rehabilitation, and infection is the number one cause of death
in stroke after day 1. It has become clear that CNS injury is
an independent risk factor, which specifically and signifi-
cantly increases susceptibility to infection. Only recently it
has been realized that CNS injury, including stroke, induces
immunodepression, and that this is the mechanism by which
CNS injury leads to secondary immunodeficiency (CNS
injury-induced immunodepression [CIDS]7) and infection.
Results from our laboratory and other groups have explored
the mechanisms how stroke downregulates peripheral immu-
nity (Figure 1). In brief, within 3 days after focal cerebral
ischemia, mice develop spontaneous pneumonia and septice-
mia. Focal cerebral ischemia induces an extensive apoptotic
loss of lymphocytes and a shift from T helper cell (Th)1 to
Th2 cytokine production. Secondary lymphatic organs like
spleen and thymus atrophy after focal cerebral ischemia.8
In parallel to the changes in the adaptive immune system,
monocyte counts and function are compromised as well.

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015


Dirnagl et al Stroke-Induced Immunodepression 772
brain. Lateralization of the structures of the vegetative ner-
vous system might also serve to explain some localization-
administration of IFN at day 1 after focal cerebral ischemia
greatly decreases the bacterial burden. Importantly, the IFN
deficiency and the occurrence of bacterial infections can be
prevented by blocking the sympathetic nervous system, but
not the hypothalamo-pituitary-adrenal axis. Furthermore, ad-
ministration of the -adrenoreceptor blocker propranolol
drastically reduces mortality after middle cerebral artery
occlusion.9 Immunodepression after stroke can be detected
within a few hours after induction of ischemia, and lasts for
several weeks. These and other studies indicate that a cate-
cholamine-mediated defect in early lymphocyte activation is
the key factor in the impaired anti-bacterial immune response
after stroke.

Is Stroke-Induced Pneumonia a Result of


Aspiration, Immunodepression, or Both?
Bacterial pneumonia is the most common cause of death in
acute stroke patients. Reduction of bulbar reflexes, drowsi-
ness, dysphagia, and subsequent aspiration are considered to
be major contributors to the high incidence of bacterial
pneumonia after stroke.10 In a murine model of aspiration
pneumonia, we have evaluated whether stroke-induced im-
munodepression contributes to the development of pneumo-
nia after stroke. We were able to show that intranasal
aspiration of only 200 colony-forming units of Streptococcus
pneumoniae cause severe pneumonia and bacteremia in mice
after transient middle cerebral artery occlusion. In contrast,
200 000 colony-forming units are needed to induce pneumo-
nia of similar severity, but fail to induce bacteremia in sham
animals. Aspiration pneumonia in animals after focal cerebral
ischemia was prevented by -adrenoceptor blockade, sug-
gesting that immunodepression by sympathetic hyperactivity
is essential for poststroke pneumonia.11 Therefore, the dele-
terious combination of stroke-facilitated aspiration and
stroke-induced immune deficiency drastically increases the
susceptibility to infection.

What Causes
Stroke-Induced Immunodepression?
Although the phenomenon of stroke-induced immunodepres-
sion is well-established, it remains unclear which signals and
mechanisms trigger the sympathetic nervous system and the
hypothalamic–pituitary axis to downregulate immune re-
sponses after brain ischemia. Several lines of clinical and
experimental evidence indicate that pro-inflammatory cyto-
kines produced by damaged brain tissue can directly lead to
hypothalamic–pituitary axis and CNS activation. Increased
levels of cytokines, such as interleukin-1 , tumor necrosis
factor- , and interleukin-6, have been measured after stroke
in brain parenchyma and cerebrospinal fluid. Because the
autonomic system of the CNS is “hard-wired” with secondary
lymphoid organs, interruption of these circuits can result in
immune dysfunction. Stroke can lead to direct damage of
sympathetic CNS structures involved in vegetative neuroim-
munomodulation. Further support for the concept of an at
least partially neurogenic nature of CIDS comes from studies
on the lateralization of the autonomic nervous system in the

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015


772 Stroke February 2007 Part II
ment with various types of stem or progenitor cells can
favorably improve outcome after experimental stroke. For
dependent effects of neural-immune interactions subsequent example, hematopoietic or mesenchymal stem cells, even
to stroke.7 Therefore, multiple causes—“unspecific” stress
response, CNS injury-specific neurogenic signaling, and local
CNS inflammation— have to be considered as triggers of
systemic immunodepression.

Does Immunodepression After Stroke Protect


the Brain?
At least in animal experiments it has been demonstrated that
stroke-induced immunodepression underlies the increased
rate of infection and unfavorable outcome after focal cerebral
ischemia.9 In principal, immunomodulatory therapy (eg,
IFN , granulocyte colony-stimulating factor, thymopentin)
to reverse the immunodepression after stroke might seem to
be a straightforward strategy to reduce the negative conse-
quences of CIDS. However, too little is known about CIDS to
rule out that it is an adaptive response, which is CNS
protective at the price of an increased risk of infection. For
example, it has been demonstrated that T lymphocytes and
IFN contribute to the inflammatory and thrombogenic brain
injury that results from cerebral ischemia.12 Therefore, deple-
tion of circulating T-cell populations and suppression of
IFN expression, which is the key mechanisms of stroke-
induced immunodepression, might counteract the inflamma-
tory brain after stroke.7 Indeed, recent evidence from our
laboratory indicates that stroke-induced immunodepression
suppresses autoaggressive Th1 responses. The blood– brain
barrier disruption and tissue destruction after stroke expose
brain epitopes that are normally “invisible” to the immune
system. This may induce autoregulatory, anti-inflammatory,
and potentially regenerative T-cell responses,13 or rather
prime the immune system to attack the CNS that may be a
particular problem in the context of systemic inflammatory
response14 or a second ischemic event.
In a mouse model of focal cerebral ischemia we detected
increased numbers of myelin oligodendrocyte glycoprotein
(MOG) specific T-cells in the spleen, and an increased influx
of MOG-specific T-cells into the brain. After experimental
stroke, mice transgenic for a MOG-specific T-cell receptor
developed clinical signs of mild experimental autoimmune
encephalitis, which is the animal model of multiple sclerosis.
Transfer of T-cells of MOG–T-cell receptor transgenic mice
induced experimental autoimmune encephalitis in naive mice.
Adoptive transfer of wild-type splenocytes (which were
harvested after focal cerebral ischemia) before induction of
focal cerebral ischemia worsens outcome, whereas transfer of
naive splenocytes has no effect (manuscript in preparation).
These findings indicate that stroke triggers an autoreactive
phenotype, against which stroke-induced immunodepression
may partially protect (Figure 2). Clearly, further research is
needed before immunomodulation should be considered in
the treatment of human stroke.

Cell Therapy and the Immune System in


Stroke: A Hypothesis
Recently, a number of groups have demonstrated that treat-

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015


analyzed in the intention-to-treat population and 66 in the
per-protocol population. In the intention-to-treat analysis,
patients receiving moxifloxacin showed a strong tendency
toward a lower infection rate compared with patients receiv-
ing placebo. Per-protocol analysis revealed a significant

Figure 2. Hypothesis of adaptive function of stroke induced


immunodepression.

when given via a systemic route, seem to foster recovery.15


Most of the research in this field has concentrated on the
homing of these cells toward the ischemic lesion, and their
putative differentiation to neuronal phenotypes. However,
autologous, heterologous, or xenologous transplantation have
multiple and complex effects on the immune system. These
may be the result of the presentation of foreign epitopes (even
in autologous transplantation, because cells are often culti-
vated in the presence of sera containing xenologous growth
factors), the apoptotic death of a substantial number of
transplanted cells,16 or endogenous production of immuno-
modulators by stem cells (eg, indoleamine17). Because
long-term outcome after stroke is heavily modulated by the
immune system, we hypothesize that the recently discovered
effects of cell therapy in stroke are at least partially the result
of its action on the immune system of the host. Clearly,
further research is needed to clarify this issue.

How to Prevent the Negative Consequences


of Immunodepression?
Because infection, particularly pneumonia, appears to be the
clinically most significant consequence of stroke-induced
immunodepression, and because immunomodulation at pres-
ent is not a viable therapeutic option, preventive antibiotic
therapy has been proposed as a measure to improve outcome
after stroke. In an animal model of stroke, prophylaxis against
bacterial infection dramatically improved mortality and re-
duced infarct sizes.18 In a recently published randomized
clinical trial (ESPIAS trial19), the fluochinolone levofloxacin
did not prevent infections in patients with acute stroke. In
the PANTHERIS trial (http://controlled-trials.com/isrctn/
trial/PANTHERIS/0/74386719.html), we are currently test-
ing whether another fluochinolone, moxifloxacin, prevents
infections in patients with severe stroke. PANTHERIS is a
double-blind, randomized, controlled phase IIb multicenter
trial in which 80 patients with severe stroke (National
Institutes of Health Stroke Scale 11) of the middle cerebral
artery territory were randomized to receive moxifloxacin
(400 mg intravenous daily) or placebo for 5 days starting
within 36 hours after stroke onset. Seventy-nine patients were

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015


Dirnagl et al Stroke-Induced Immunodepression 773

reduction of the cumulative infection rate (mainly pneumo- 2. Hart J. Inflammation. 1: Its role in the healing of acute wounds. J Wound
nia) in the moxifloxacin compared with the placebo group. Care. 2002;11:205–209.
3. Dirnagl U. Inflammation in stroke: the good, the bad, and the unknown.
Patients with infectious complications showed strong signs Ernst Schering Res Found Workshop. 2004;87–99.
of systemic immunodepression (monocyte deactivation, T-cell 4. Chamorro A, Hallenbeck J. The harms and benefits of inflammatory and
dysfunction, etc) before clinical onset of infection. Our data immune responses in vascular disease. Stroke. 2006;37:291–293.
5. Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci.
(manuscript in preparation) indicate that preventive antibac- 2004;5:146 –156.
terial therapy with moxifloxacin prevents infectious compli- 6. Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, Wang X,
cations in patients with severe stroke. In addition, inhibition Lo EH. Role of matrix metalloproteinases in delayed cortical responses
after stroke. Nat Med. 2006;12:441– 445.
of cellular immune function after acute stroke appears to 7. Meisel C, Schwab JM, Prass K, Meisel A, Dirnagl U. Central nervous
increase the risk of infectious complications. The conflicting system injury-induced immune deficiency syndrome. Nat Rev Neurosci.
results of the ESPIAS and PANTHERIS may be explained by 2005;6:775–786.
8. Offner H, Subramanian S, Parker SM, Wang C, Afentoulis ME, Lewis A,
patient selection, differences in antibiotics, dosages, and Vandenbark AA, Hurn PD. Splenic atrophy in experimental stroke is
duration of treatment. Larger trials are needed to characterize accompanied by increased regulatory T cells and circulating macro-
stroke-induced immunodepression in humans and to answer phages. J Immunol. 2006;176:6523– 6531.
the questions whether immunodepression and consequently 9. Prass K, Meisel C, Hoflich C, Braun J, Halle E, Wolf T, Ruscher K,
Victorov IV, Priller J, Dirnagl U, Volk HD, Meisel A. Stroke-induced
infection have a clinical impact that can be positively affected immunodeficiency promotes spontaneous bacterial infections and is
by preventive anti-infective therapy. mediated by sympathetic activation reversal by poststroke T helper cell
type 1-like immunostimulation. J Exp Med. 2003;198:725–736.
10. Perry L, Love CP. Screening for dysphagia and aspiration in acute stroke:
Conclusions a systematic review. Dysphagia. 2001;16:7–18.
There is emerging experimental and clinical evidence that 11. Prass K, Braun JS, Dirnagl U, Meisel C, Meisel A. Stroke propagates
brain–immune interactions play an important role for out- bacterial aspiration to pneumonia in a model of cerebral ischemia. Stroke.
come after stroke. Because these interactions may have 2006; 37:2607–2612.
12. Yilmaz G, Arumugam TV, Stokes KY, Granger DN. Role of T lym-
protective, destructive, or regenerative effects in the brain, phocytes and interferon-gamma in ischemic stroke. Circulation. 2006;
and also impact the organism as a whole, development of 113:2105–2112.
therapeutic strategies is not straightforward. Further research 13. Schwartz M, Kipnis J. Protective autoimmunity and neuroprotection in
inflammatory and noninflammatory neurodegenerative diseases. J Neurol
is needed to understand the signals and mechanisms by which Sci. 2005;233:163–166.
the adaptive and innate immune systems react to brain tissue 14. Becker KJ, Kindrick DL, Lester MP, Shea C, Ye ZC. Sensitization to
damage, and to unravel the consequences that this has for the brain antigens after stroke is augmented by lipopolysaccharide. J Cereb
Blood Flow Metab. 2005;25:1634 –1644.
patient. 15. Lindvall O, Kokaia Z. Recovery and rehabilitation in stroke: stem cells.
Stroke. 2004;35:2691–2694.
Sources of Funding 16. Thum T, Bauersachs J, Poole-Wilson PA, Volk HD, Anker SD. The dying
This study was supported by the Hermann and Lilly Schilling stem cell hypothesis: immune modulation as a novel mechanism for
Foundation and the Helmholtz-Gemeinschaft. progenitor cell therapy in cardiac muscle. J Am Coll Cardiol. 2005;46:
1799 –1802.
17. Barry FP, Murphy JM, English K, Mahon BP. Immunogenicity of adult
Disclosures mesenchymal stem cells: lessons from the fetal allograft. Stem Cells Dev.
U.D., C.M., K.P., and A.M. are inventors on a patent application 2005;14:252–265.
(owner: Charité-Humboldt University) in which stroke-induced im- 18. Meisel C, Prass K, Braun J, Victorov I, Wolf T, Megow D, Halle E, Volk
munodepression is claimed. HD, Dirnagl U, Meisel A. Preventive antibacterial treatment improves the
general medical and neurological outcome in a mouse model of stroke.
References Stroke. 2004;35:2– 6.
1. Redd MJ, Cooper L, Wood W, Stramer B, Martin P. Wound healing and 19. Chamorro A, Horcajada JP, Obach V, Vargas M, Revilla M, Torres F,
Cervera A, Planas AM, Mensa J. The Early Systemic Prophylaxis of
inflammation: embryos reveal the way to perfect repair. Philos Trans R
Infection After Stroke study: a randomized clinical trial. Stroke. 2005;
Soc Lond B Biol Sci. 2004;359:777–784.
36:1495–1500.

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015


Stroke-Induced Immunodepression: Experimental Evidence and Clinical Relevance
Ulrich Dirnagl, Juliane Klehmet, Johann S. Braun, Hendrik Harms, Christian Meisel, Tjalf
Ziemssen, Konstantin Prass and Andreas Meisel

Stroke. 2007;38:770-773
doi: 10.1161/01.STR.0000251441.89665.bc
Stroke is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2007 American Heart Association, Inc. All rights reserved.
Print ISSN: 0039-2499. Online ISSN: 1524-4628

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://stroke.ahajournals.org/content/38/2/770

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Stroke can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office.
Once the online version of the published article for which permission is being requested is located, click
Request Permissions in the middle column of the Web page under Services. Further information about this
process is available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Stroke is online at:
http://stroke.ahajournals.org//subscriptions/

Downloaded from http://stroke.ahajournals.org/ by guest on May 24, 2015

Vous aimerez peut-être aussi