You are on page 1of 8

Molecular and Cellular Neuroscience 53 (2013) 26–33

Contents lists available at SciVerse ScienceDirect

Molecular and Cellular Neuroscience


journal homepage: www.elsevier.com/locate/ymcne

The effect of stroke on immune function☆,☆☆


Roberta Brambilla a, 1, Yvonne Couch b, 1, Kate Lykke Lambertsen c,⁎, 1
a
The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
b
The Department of Pharmacology, University of Oxford, Oxford, UK
c
The Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark

a r t i c l e i n f o a b s t r a c t

Article history: Neurological disorders affect over one billion lives each year worldwide. With population aging, this number is
Received 15 March 2012 on the rise, making neurological disorders a major public health concern. Within this category, stroke represents
Accepted 22 August 2012 the second leading cause of death, ranking after heart disease, and is associated with long-term physical disabil-
Available online 30 August 2012
ities and impaired quality of life.
In this review, we will focus our attention on examining the tight crosstalk between brain and immune
Keywords:
Stroke
system and how disruption of this mutual interaction is at the basis of stroke pathophysiology. We will
Immune responses also explore the emerging literature in support of the use of immuno-modulatory molecules as potential
Autonomic nervous system therapeutic interventions in stroke. This article is part of a Special Issue entitled 'Neuroinflammation in
HPA axis neurodegeneration and neurodysfunction'.
© 2012 Elsevier Inc. All rights reserved.

Introduction Risk factors for developing stroke include co-morbid diseases such
as atherosclerosis, obesity, diabetes, hypertension and peripheral infec-
Ischemic stroke results in a multitude of CNS events characterized tion (Emsley et al., 2008; Hankey, 2006). Common to all is the associa-
ultimately by neuronal and glial cell death, and is also marked by tion with elevated systemic inflammation, which increasing evidence
numerous peripheral events including cardiovascular, endocrine and points at having a causative role in the development of these diseases
immune dysregulation (Emsley et al., 2008; Stevens and Nyquist, (Hansson and Libby, 2006). Several clinical studies have reported
2007). The contribution of the immune system to the development more severe neurological deficits in stroke patients with preceding
and progression of cerebral infarcts is well established. However, re- infection (McColl et al., 2009). Furthermore, elevated systemic concen-
cent evidence suggests that it may also contribute to recovery and re- trations of a number of inflammatory markers have been associated
pair in the long term after ischemic damage (Gelderblom et al., 2009; with stroke incidence (Rodriguez-Yanez et al., 2008), emphasizing
Hug et al., 2009). Stroke patients face severe immunological chal- the role of inflammatory events occurring outside the brain prior to,
lenges while still in intensive care units, so much so that the most during and after stroke, on stroke susceptibility and outcome.
common, fatal, post-stroke complication is pneumonia (Aslanyan et Even though these pre-existing conditions are major contributors
al., 2004; Johnston et al., 1998; Katzan et al., 2003). Indeed, a recent to stroke incidence and physiopathology, this review will specifically
meta-analysis has revealed that infection after acute ischemia can focus on the direct effects of stroke on peripheral immune function,
complicate recovery in up to 30% of cases (Westendorp et al., 2011). since dysregulation of such immune response has clear negative im-
While in the past the assumption was that post-stroke infections were plications on patient outcome, and a better understanding of these
dependent on pre-existing co-morbidities and mismanagement of pa- events is critical in devising appropriate and comprehensive thera-
tient care, it is now clear that post-stroke immunodepression repre- peutic strategies for the treatment of stroke patients. The effect of in-
sents an independent factor associated with increased susceptibility to flammation on stroke outcome will be covered separately by Stuart
infections (Emsley et al., 2008). Allan on the review dealing with the afferent pathways in stroke.

Stroke and central inflammation


☆ The authors have no conflict of interest.
☆☆ The authors would like to acknowledge financial support from the Lundbeck
Foundation and the Danish Medical Research Council to Dr. Kate Lykke Lambertsen Since the brain has a very high glucose and oxygen demand, distur-
and from The Miami Project to Cure Paralysis to Dr. Roberta Brambilla. bances in the blood supply to the brain rapidly lead to the depletion of
⁎ Corresponding author at: Department of Neurobiology Research, Institute of Molecular these substrates and the development of an ischemic infarct with ac-
Medicine, University of Southern Denmark, J.B. Winsløwsvej 21, st., DK-5000 Odense C,
Denmark.
companying necrosis of neurons, glial cells and small vessels within
E-mail address: klambertsen@health.sdu.dk (K.L. Lambertsen). the affected territory. Depletion of cellular energy supplies (such as
1
All three authors contributed equally to this work. adenosine triphosphate (ATP)) occurs within minutes, resulting in the

1044-7431/$ – see front matter © 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.mcn.2012.08.011
R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33 27

accumulation of lactic acid within the tissue. Ischemia also leads to the post-stroke infections is directly correlated with the magnitude of
formation of free radicals that can induce cell damage, and to increased the stroke itself, and specifically with how extensive the infarct area
release of excitatory glutamate (for recent review see (Iadecola and is (Hug et al., 2009). The size of the infarct area also parallels the se-
Anrather, 2011)). ATP depletion and glutamate release result in verity of leukocytopenia, which directly correlates with stroke-
uncontrolled calcium-ion influx into the cells leading to activation of in- associated immunodepression. The occurrence of leukocytopenia fol-
tracellular lipases and proteolytic enzymes and, ultimately, destruction lowing stroke has been well documented in patients, with reports
of the cell. The ability of glutamate to kill neurons by excessive activation dating back over 40 years (Czlonkowska et al., 1979). Rapid reduction
of glutamate receptors is referred to as excitotoxicity (Mergenthaler et of lymphocyte counts and functional deactivation of monocytes and
al., 2004). The early excitotoxicity induced by the local energy deficit T helper type 1 cells have been observed in acute stroke patients, with
causes fast necrotic cell death in the core area of the infarct (Lipton, more pronounced immunodepression in patients with severe clinical
1999). The ischemic penumbra that surrounds the infarct core suffers deficit or large infarction (Haeusler et al., 2008). Lymphocytopenia
milder damage, partly due the numerous collaterals and anastomoses, does correlate with the occurrence of post-stroke infections (Haeusler
which supply the neurons within the penumbra (Astrup et al., 1981). et al., 2008; Hug et al., 2009). In some instances, rather than a generalized
This area is characterized by compromised blood flow, impaired neuro- reduction in total lymphocyte counts, only selective lymphocytopenia in
nal functionality, but preserved structural integrity (Astrup et al., 1981). the NK cell subset was reported immediately after stroke (Hug et al.,
In addition, astrocytes are more resistant to cerebral ischemia than neu- 2009).
rons and react to hypoxia by upregulating their glycolytic capacity The tight relationship between lymphocytopenia, size of infarct
allowing a continued uptake of glutamate from the synaptic cleft in and the occurrence of post-stroke infections has been demonstrated
the penumbral area (Marrif and Juurlink, 1999). It has been observed also in animal models of stroke. In mice, severely reduced lymphocyte
that the penumbra has suppressed cortical protein synthesis, but pre- counts (B cells and CD4 + T helpers, especially) were found in lym-
served ATP content (del Zoppo et al., 2011; Hossmann, 2006). For phoid organs (spleen and thymus) within 12 h after stroke (Prass et
these reasons the penumbral area is still potentially salvageable and, al., 2003). This was paralleled by spontaneous bacteremia and pneu-
thus far, has been the target of stroke therapy (del Zoppo et al., 2011). monia (often leading to death), which were completely prevented
After ischemia, resident cells, including microglia and astrocytes, by administration of a sympathetic blocker, but not by inhibition of
are quickly activated and circulating leukocytes are recruited to the the hypothalamic-pituitary-adrenal (HPA) axis (the paravetricular
ischemic lesion. It is believed that, early on, endogenous signals nucleus in the hypothalamus, the anterior pituitary gland and the cor-
such as damage-associated molecular patterns (DAMPs; i.e. heat tex of the adrenal glands), suggesting that a catecholamine-mediated
shock protein (HSP)60, HSP70 and high-mobility-group box-1 defect in early lymphocyte activation is the key factor in the impaired
(HMGB1)) are released from stressed and dying cells and subse- antibacterial immune response after stroke (Prass et al., 2003). A re-
quently bind to toll-like receptors (TLRs), especially TLR2 and TLR4, cent study by Wong and colleagues has highlighted the role of invari-
located on resident microglia and astrocytes, resulting in downstream ant natural killer T (iNKT) cells in the defense against post-stroke
activation of MyD88- and/or TRIF-dependent pathways leading to ac- infections (Wong et al., 2011). Modulation of hepatic iNKT through
tivation of nuclear factor kappa B- and/or IRF3-dependent gene tran- blockade of noradrenergic neurotransmitters or directly with admin-
scription (for a thorough review on this topic, please refer to Marsh et istration of β-galactosylceramide results in reduced infection and as-
al. (2009)). This triggers the synthesis of primarily microglia-derived sociated lung injury after stroke, demonstrating that these cells act as
pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6 and conductor of immunity, meaning that their acute responses modulate
tumor necrosis factor (TNF) (reviewed in (Lambertsen et al., 2012)), and facilitate the adaptive immune response (Wong et al., 2011).
chemokines (CC and CXC chemokines) (Mirabelli-Badenier et al., Because of the correlation between post-stroke infections and
2011), nitric oxide and reactive oxygen species, which, when present clinical outcome, the prophylactic use of antibiotics to prevent such
at high levels, can exacerbate cell death and cause break down of the infections and improve the outcome has been proposed. The data
blood–brain barrier (BBB) (for recent review see (Iadecola and emerging from clinical studies, however, are contradictory. For exam-
Anrather, 2011)). Cytokines and chemokines also induce the ple, no benefit was found with prophylactic administration of the flu-
upregulation of adhesion molecules on the vascular endothelium, fa- oroquinolone levofloxacin, a broad-spectrum antibiotic, in acute
voring diapedesis of circulating leukocytes that may further contrib- stroke patients admitted within 24 h after symptom onset. The rate
ute to brain injury. of stroke-related infections at 7 days was identical to the placebo
group, and levofloxacin administration was directly correlated with
Stroke-associated infection poor clinical outcome (Chamorro et al., 2005). On the other hand,
prophylactic treatment with another fluoroquinolone, moxifloxacin,
Post-stroke infections represent one of the principal complications resulted in the significant reduction of post-stroke infections, al-
adversely affecting the clinical outcome in stroke patients. Although though not in the improvement of the clinical outcome (Harms et
some infections may occur as a direct consequence of dysphagia caus- al., 2008). This is in contrast with data obtained in a mouse model
ing aspiration, or may be linked to the advanced age of the patient, it of middle cerebral artery occlusion (MCAO), where moxifloxacin, a
is increasingly apparent that stroke itself represents a risk factor for similar broad-spectrum antibiotic, administration significantly re-
infections due to the induction of a so-called post-stroke immuno- duced infarct size (Bao et al., 2010). A possible explanation for the
depression syndrome, which occurs immediately after stroke failure of these molecules in human therapy is the neurotoxicity of
(Chamorro et al., 2007; Vermeij et al., 2009). Indeed, the fact that fluoroquinolones, which could be offsetting their beneficial antimi-
the majority of post-stroke infections manifest within three days of crobial effect. In contrast, other classes of broad-spectrum antibiotics
hospitalization is further indication that immunodepression is in- (e.g. penicillins and tetracyclines) have shown protective effects. In-
volved, and infections are not simply a secondary outcome related deed, prophylactic administration of the broad spectrum semisyn-
to patient care (Westendorp et al., 2011). A recent meta-analysis of thetic penicillin mezlocillin in combination with the beta-lactamase
the 87 clinical studies conducted thus far, where rates of post-stroke inhibitor sulbactam decreased incidence and severity of fever within
infections were examined, has indicated that infection complicated the first 3–4 days after stroke and was associated with a lower rate
acute stroke in 30% of patients, with rates varying considerably be- of post-stroke infection and improved long term outcome (Schwarz
tween 5 and 67% (Westendorp et al., 2011). Pneumonia and urinary et al., 2008). Finally, minocycline, a semisynthetic second generation
tract infections each occurred in 10% of patients, with pneumonia sig- tetracycline, is the antibiotic that perhaps holds the highest promise.
nificantly associated with death. It is also clear that the severity of After being proven effective in numerous experimental models of
28 R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33

stroke (Hayakawa et al., 2008; Hewlett and Corbett, 2006; Liu et al., Hepatic signaling pathways
2007), an open-label placebo-controlled clinical trial found that Very little is known about the pathways responsible for turning on
minocycline treatment significantly improved the clinical outcome, the genes of the APR in the liver after brain injury. Below, we will re-
even though it did not protect from post-stroke infections. This view those known to be involved in the activation of the HPA axis,
seems to suggest that the protective effect of minocycline in stroke and the autonomic nervous system (both sympathetic and parasym-
may not be dependent on its antibacterial properties, but rather on pathetic) as well as emerging putative mechanisms.
its anti-inflammatory effect, which has been associated with a de- In terms of neuronal activation, CNS injuries have previously been
crease in microglial activation and nitric oxide production, inhibition shown to ‘switch-on’ neuronal pathways, i.e. initiate neurotransmis-
of matrix metalloproteinases (MMPs) (e.g. MMP2, MMP9), and inhi- sion. This CNS activation has been shown to be mediated by choliner-
bition of apoptotic cell death (Zemke and Majid, 2004). Recently, in gic efferents to the liver from the brain and can be attenuated by vagal
a small exploratory trial minocycline was found to be safe and well tol- nerve lesion (Ottani et al., 2009). This suggests that the brain is
erated in intravenous doses, either given alone or in combination with switching on neurotransmission in order to signal injury to the pe-
tissue plasminogen activator (tPA) (Fagan et al., 2010), supporting the riphery. This relatively recent idea has been dubbed ‘central neuro-
rationale for a possible clinical application in stroke. genic neuroprotection’ and suggests the existence of both central
Studies in animal models have also underscored that post-stroke adrenergic and cholinergic circuits within the brain that protect neu-
infections may be effectively prevented with the use of immunomod- rons from the aftermath of CNS injury, and is therefore particularly
ulatory molecules (Prass et al., 2003; Wong et al., 2011), rather than relevant in stroke research (Feinstein et al., 2002; Galea et al., 2003).
antibiotics, and this approach could be explored in clinical studies, The role of the autonomic nervous system in the communication
as it would allow to bypass potential downfalls of prolonged antimi- between brain and periphery has been described by a number of
crobial therapy, namely the high incidence of bacterial resistance. studies. As for the PNS, peripheral vagal stimulation significantly re-
duces lesion volume in a rat model of stroke (Hayakawa et al.,
2008). As for the SNS, administration of clenbuterol, a brain penetrant
Brain-periphery signaling post-stroke β-adrenergic agonist, shows protection in animal models of
excitotoxicity, demonstrating both the anti-inflammatory potential
The activation of the CNS after ischemia results in signaling to pe- of the autonomic nervous system and the concept of neurogenic
ripheral organs, particularly the gut and the liver, which in turn elicit neuroprotection (Ryan et al., 2011). In addition, studies show that
a substantial immune response. This concept is relatively new and changes in infusion rates from the left or right carotid arteries deter-
largely unexplored, and the signaling pathways that mediate these mine whether tachycardia (sympathetic activation) or bradycardia
events are mostly unknown. Significant delays are known to exist (parasympathetic activation) will develop. Patients with right-sided
between the central challenge and the peripheral inflammatory re- stroke affecting the insular cortex most frequently show tachycardia,
sponse (Blond et al., 2002). Interestingly, in ischemia, the peak central suggesting an increase in sympathetic activation (Colivicchi et al.,
inflammatory phase has been shown to be induced at approximately 2004; Tokgozoglu et al., 1999).
24 h post-ischemia, whereas the peak peripheral inflammatory phase The autonomic nervous system is also capable of directly stimulat-
occurs as early as 4 h post-ischemia (Chapman et al., 2009). This pe- ing immune cells. Indeed, adrenergic receptors such as the
ripheral inflammation is characterized by an acute phase response β-adrenoceptor, are expressed on T helper type-1 (Th1) lymphocytes,
(APR) in the liver, where it is reported that close to one thousand B cells and macrophages (Kohm and Sanders, 2001; Mackroth et al.,
genes are switched on in response to CNS injury (Campbell et al., 2011). In the periphery, catecholamines, such as noradrenaline, are
2007). released during stress, a common phenomenon after acute stroke
One mechanism by which the brain communicates with the pe- (Anne et al., 2007; Oto et al., 2008). Therefore, it is not unreasonable
ripheral immune system is via the HPA axis in concert with the auto- to postulate that systemic release of cytokines after stroke might be
nomic nervous system (Fig. 1). Interestingly, cytokines such as TNF, mediated, in part, by the autonomic nervous system and catechol-
IL-1β and IL-6 are pivotal in the cross talk between brain and immune amines (Marz et al., 1998). Both locally produced cytokines in the
system. Indeed, they activate the autonomic nervous system and the brain, and direct brain stem irritation (by local cytokines or compres-
HPA axis, where release of catecholamines and glucocorticoids can sion) can trigger strong sympathetic activation and release of cate-
modulate the immune function (Turnbull and Rivier, 1999). It is be- cholamines which, in turn, can lead to systemic release of cytokines
lieved that cytokines known to be significantly increased after stroke, (Marz et al., 1998; Woiciechowsky et al., 1998, 1999). While these
such as IL-1 (Clausen et al., 2005), act on the hypothalamus (Haddad studies have shown that the activation of the SNS is capable of pro-
et al., 2002; Uehara et al., 1987) leading to release of corticotropin re- found immunomodulation, there remains some confusion over the
leasing hormone (CRH) into the hypophyseal portal blood supply. impact this will have on outcome, some maintain that inhibition of au-
Here CRH stimulates the secretion of adrenocorticotropic hormone tonomic signaling can produce beneficial outcomes after stroke
(ACTH) from the anterior pituitary gland (Anne et al., 2007), which (Savitz et al., 2000). The direct pathways of communication between
in turn circulates to the adrenal glands. Any imbalance in the homeo- the injury site and the SNS immediately post-stroke are currently
stasis of this system caused by cerebral ischemia may result in activa- speculative. However, it is clear that some uncoupling occurs be-
tion of the HPA axis, causing increased production of glucocorticoids tween the CNS, SNS and their control of the immune system during
from the adrenal glands (Krugers et al., 1995). Since immune cells ex- the immediate post-stroke period.
press receptors for hormones and neurotransmitters (Heijnen, 2007), The main target of PNS pathways, such as output from the vagus,
it is believed that the HPA axis, via glucocorticoid release, can lead to is the nicotinic acetylcholine receptor α7 subunit (nAChRα7), which
lymphopenia and lymphocyte dysfunction, particularly monocyte de- is expressed on both neurons and resident immune cells such as mac-
activation (Fig. 1) potentially resulting in bacteremia and pneumonia rophages and microglia (Shytle et al., 2004). This affects inflammation
in stroke patients (reviewed in (Prass et al., 2003)). Furthermore, via the nuclear factor kappa B (NF-κB) pathway (Borovikova et al.,
overstimulation of both the sympathetic (SNS) and parasympathetic 2000). Stimulation of the peripheral immune system of vagotomized
(PNS) nervous systems results in increased circulating levels of both mice has lead to the conclusion that expression of nAChRα7 on
catecholamines and acetylcholine (Ach) (Anne et al., 2007; Elenkov Kupffer cells enables the hepatic branch of the vagus to suppress
et al., 2000; Franceschini et al., 1994) (Fig. 1). The detrimental effects the production of reactive oxygen species in these cells (Hiramoto
of these neurotransmitters on the peripheral immune system is et al., 2008). This ‘top down’ immune suppression has been confirmed
discussed in detail below. in nAChRα7 knockout mice, which show significantly elevated levels
R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33 29

Fig. 1. Schematic representation of the communication pathways between the stroke-injured brain and the peripheral immune system. Pro-inflammatory cytokines stimulate neurons in
the paraventricular nucleus of the hypothalamus to secrete CRH, which then facilitates the release of ACTH from the anterior pituitary. This leads to the release of GCs from the adrenal
cortex resulting in suppression of the production of pro-inflammatory mediators and facilitates the release of anti-inflammatory mediators resulting in a classic negative feedback loop.
The SNS also plays an important role in the communication between the stroke injured brain and the peripheral immune system. Activation of the SNS causes the release of CAs from sym-
pathetic nerve terminals and from the adrenal medulla resulting in inhibition of Th1 pro-inflammatory activities, giving way to the predominance of Th2 anti-inflammatory activities. Also
activation of the parasympathetic nervous system is believed to play an important role in the communication between the injured brain and the immune system. Activation via the vagus
nerve, the cholinergic anti-inflammatory pathway, results in release of ACh acting on cholinergic receptors on macrophages leading to a decrease in the production of anti-inflammatory
cytokines. ACTH, adrenocorticotrophic hormone; APR, acute phase response; BBB, blood brain barrier; CAs, catecholamines; CRH, corticoreleasing hormone; GCs, glucocorticoids; HPA,
hypothalamic-pituitary-adrenal; IL, interleukin; IL-1Ra, interleukin-1 receptor antagonist; Mф, macrophages; NA, noradrenaline; NK, natural killer; SNS, sympathetic nervous system;
TGFβ, transforming growth factor beta; Th, T helper cells; TNF, tumor necrosis factor.

of TNF and IL-6 in response to immune challenge (Fujii et al., 2007). central inflammatory diseases such as cerebral malaria have shown
Oxygen deprivation in nAChRα7 knockout animals results in in- that MPs can be used as markers of cerebral dysfunction (Pankoui
creased damage, suggesting that this receptor is responsible not Mfonkeu et al., 2010). However, data from acute stroke patients
only for the peripheral inflammatory response, but also for regulating have been less promising so far (Williams et al., 2007) since no differ-
inflammatory output within the CNS (Egea et al., 2007). These data ences were found in MP characteristics. While these studies could not
suggest that central activation of the PNS by stroke will largely de- distinguish between stroke and stroke-mimic patients (those who
press the immune system, whereas activation of the SNS will largely present the symptoms of a stroke but do not show any underlying
activate it. vascular anomalies), in other instances the phenotype of MPs has
One potential, and novel, mechanism of inflammatory output from been successfully used to discriminate between cerebrovascular
the CNS is the microparticles (MPs). These are membrane-derived events (Haeusler et al., 2008). While work studying MPs and their po-
vesicles produced as a result of cell stress and can be released through tential as a communication pathway between the CNS and the periph-
blebbing from a variety of different cell types including neutrophils, ery is currently in its infancy, the promising studies using central
endothelial cells and microglia. These microparticles would theoreti- inflammatory diseases such as cerebral malaria pave the way for inter-
cally be small enough to cross an intact blood brain barrier and thus esting future work.
injury in the CNS would be able to use them as a mechanism of com-
municating with the periphery. Recent advances have allowed MPs to Brain–gut axis
be phenotyped, showing not only the cellular origin of the MPs but The gut is composed of three key immunological components: the
also distinct populations after specific types of injury. Such phenotyp- gut epithelium (the first point of contact for foreign bodies), the mucosal
ic differences may suggest a mechanism of discreet communication immune system (a particularly sensitive site high in IgA-positive cells),
between the CNS and the peripheral immune system. Studies in and the gut microflora (commensal bacteria that live in a symbiotic
30 R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33

relationship with the host). Within the mucosal immune system, reduction in activated microglia and infiltrating macrophages
lymph drainage occurs at Peyer's patches, sites where high numbers (Ajmo et al., 2008). Although this approach may be useful in the
of immune cells gather. Interestingly, studies using the permanent short term, questions remain as to whether this could lead to
MCAO model of stroke in rats have demonstrated stark alterations detrimental effects in the long term, given the role of the spleen in
in the gut mucosa post-ischemia (Tascilar et al., 2010). The height normal immune function, especially in stroke patients who are
and depth of the intestinal villi, a critical factor critical for mucosal intrinsically immuno-suppressed and less equipped to fight infec-
integrity, were shown to be damaged by permanent MCAO, poten- tious complications.
tially allowing indigenous gut bacteria to migrate into otherwise Rather than ablation of the spleen, a strategy adopted by others
sterile body cavities (Tascilar et al., 2010). This work has been corrobo- consisted in replenishing the spleen with human umbilical cord
rated in animal models (Maes et al., 2008; Schulte-Herbruggen et al., blood cells (HUCBC) transfused after MCAO. HUCBC localize to the
2009), where it has been suggested that stress, and therefore possibly spleen counteracting MCAO-induced spleen atrophy, and migrate to
the HPA axis, is a significant contributing factor to bacterial transloca- the site of injury in the brain, significantly reducing infarct size
tion (Tascilar et al., 2010). (Newcomb et al., 2006; Vendrame et al., 2004). Moreover, transfused
Studies specifically investigating the intestinal immune system HUCBC appeared to switch splenic cytokine expression from a pro-
have shown decreased levels of lymphocytes in Peyer's patches fol- inflammatory (TNF, IL-1β) to an anti-inflammatory (IL-10) profile
lowing cerebral ischemia (Schulte-Herbruggen et al., 2009). These (Vendrame et al., 2004), which could be the key to the therapeutic
data are in line with numerous studies reporting changes in T and B effect of HUCBC in these experimental models of stroke.
cell numbers and functionality after stroke, which will be discussed It has been proposed that one of the mechanisms sustaining splen-
below. The exact role of lymph drainage from the intestines has yet ic activation and the release of macrophages from the spleen into cir-
to be elucidated (Newberry, 2008), but current data suggest that it culation after ischemic stroke is the activation of the SNS. Indeed,
likely provides a tolerant immune barrier between gut bacteria and besides the increase in systemic catecholamine levels released into
the systemic circulation, as well as protection against pathogenic in- the circulation from the adrenal medulla as described above, MCAO
vaders. Thus far, there are no data regarding the exact mechanisms also results in elevated catecholamine levels in the spleen through di-
of communication between the brain and the gut immune system, rect splenic innervations (Young et al., 1983). Studies by Ajmo et al.
but it seems reasonable to assume that changes, at least in circulating (2009) have shown that only blockade of α and β adrenergic recep-
lymphoid cells, would be communicated to all systemic immune sys- tors, but not spleen denervation, prevented spleen atrophy and re-
tems. However, the route by which central inflammatory events af- duced infarct volume, suggesting that it is the increased systemic
fect the mucosal epithelium is currently speculative. catecholamine level resulting from adrenal release that regulates
the splenic response after stroke.
Splenic response to stroke Collectively, these studies point at a key crosstalk between spleen
and ischemic brain beginning at the very early stages of injury. Acutely,
While little is known about brain–gut communication and the po- the spleen acts as a reservoir of pro-inflammatory immune cells
tential for physiological failure in the immediate post-stroke period, ready to be deployed into the blood immediately after stroke.
the data available regarding changes in the spleen are much more ro- These cells ultimately reach the infarct area of the brain and contrib-
bust. Since the spleen is the main ‘storage facility’ for large numbers of ute to the propagation of secondary damage. At this stage, strategies
immune cells, spleen alterations after ischemic events have the poten- aimed at containing the splenic response may be beneficial. On the
tial to induce severe perturbation of the immune function. The initial other hand, long-term suppression of the splenic response may
observation of reduced cellularity in the spleens of rats subjected to hamper physiologic immune function and limit the ability to fight
cerebral ischemia (transient MCAO) was made by Gendron et al. infections, compromising the chances of recovery of stroke patients.
(2002), who reported that in ischemic animals the total number of In conclusion, it appears clear that strategies aimed at modulating
spleen leukocytes was significantly decreased compared to sham oper- the splenic response to stroke may be extremely effective in con-
ated controls from day 2 to day 28 post-ischemia. A similar pattern of taining the propagation of secondary damage within the CNS, as
splenocyte loss was also observed in mice subjected to transient long as they are delivered in a timely fashion.
MCAO, and attributed to increased apoptotic cell death of all lympho-
cyte populations (Prass et al., 2003). Interestingly, later studies by Post-ischemia immune cell function
Offner and colleagues painted a more complex picture of the splenic re-
sponse to stroke (Offner et al., 2006a,b). They found the acute phase As discussed above, the potential for the immune system to have
after stroke (1 to 22 h) to be characterized by sustained splenocyte ac- deleterious effects on infarct volume and stroke outcome is numerous.
tivation with increased expression of pro-inflammatory cytokines and However, recent studies in both mouse models and patient cohorts in-
chemokines (e.g. TNF, IL-6, IL-2, interferon gamma (IFNγ), CXCL2, dicate that dysregulation of CNS homeostasis by ischemia can have
CXCL10), mild apoptotic death of splenocytes and limited spleen weight long-lasting effects on peripheral immunity. This dysregulation causes
loss (Offner et al., 2006a). This could help explain the increased system- a central inflammatory cascade capable of eliciting a peripheral immune
ic inflammation observed immediately after stroke, which correlates response. As mentioned previously, the peak central inflammatory peri-
with the development of increased secondary damage at the infarct od is approximately 24 h post-ischemia, whereas the peak peripheral
site in the brain. At a later post-injury time (96 h) a drastic reduction inflammatory period is approximately 4 h post-ischemia (Chapman et
in spleen weight as a consequence of reduced cellularity has been al., 2009). Discrepancies between the peak response times may allow
observed, and this was associated with massive apoptotic death of for the mobilization of peripheral immune cells, which have been
splenocytes (Shimizu et al., 1999), mostly B cells and CD4+ T effector shown to play a crucial role in infarct size (Gelderblom et al., 2009).
subsets. During stroke, disruption of the BBB allows for the influx of circulating
The early splenic response after stroke involves increased pro- peripheral immune cells such as macrophages, neutrophils and lym-
duction of pro-inflammatory mediators and the deployment of pro- phocytes, each with specific temporal patterns (Nilupul Perera et al.,
inflammatory monocytes into the blood, leading researchers to sug- 2006). There is evidence that immuno-modulatory molecules capable
gest splenectomy as a possible prophylactic intervention for cerebral of blocking or limiting the influx of these cell populations into the CNS
ischemia (Izci, 2010). Indeed, splenectomized rats display a signifi- are effective in preventing the evolution of stroke damage in animal
cantly reduced infarct volume after permanent MCAO compared to models. To this effect, administration of the sphingosine 1-phosphate
non-splenectomized rats, and this is accompanied by a significant (S1P) analog FTY720 (fingolimod), which induces depletion of
R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33 31

circulating lymphocytes by preventing their egress from the lymph T and B cells post-stroke
nodes, can reduce lesion size and improve neurological outcome after The recent interest in lymphocyte function after stroke is mainly due
stroke in mice (Czech et al., 2009). This is due to a reduced invasion of to the increased likelihood of encounters between lymphocytes and
immune cells into the lesion, in addition to a direct neuroprotective ef- CNS antigens immediately after stroke. To date, research suggests that
fect. In similar fashion, other immuno-modulatory agents affecting cell immunodepression is largely caused by lymphocytopenia, abnormally
entry in the CNS have shown effectiveness. Administration of anti- low levels of circulating lymphocytes immediately after stroke. Howev-
CD11b antibody, for example, was shown to prevent the infiltration of er, work has failed to suggest a mechanism for this phenomenon, and
polymorphonucleate cells (neutrophils and monocyte/macrophages) data in human and animal models appear to be conflicting. Hug et al.
into the ischemic tissue (Chen et al., 1994a, 1994b; Chopp et al., have shown in both humans and animal models that T-cell proliferation
1994), significantly reducing infarct size and neurological deficit. Simi- and activation ex vivo are not affected by ischemia (Hug et al., 2011).
larly, antibodies against the adhesion molecule ICAM-1 (endothelium Nevertheless, reduced T cell numbers after stroke, as well as generalized
surface) or its receptor CD18 (leukocyte surface), whose interaction is splenic atrophy, have been demonstrated in a number of studies (Hug
essential for leukocyte binding to the endothelium and subsequent et al., 2011; Offner et al., 2006b; Vogelgesang et al., 2010). It has been
transendothelial migration, showed effectiveness in limiting stroke suggested that this lymphocytopenia is due to increased apoptosis
damage and promoting functional recovery (Bowes et al., 1993; Clark within all T-cell populations (Chamorro et al., 2007). Recent work on
et al., 1991a,b; Jiang et al., 1994, 1995; Zhang et al., 1995a,b). These Tregs has shown that they have potential to provide a degree of
encouraging data in experimental models prompted the initiation immunoprotection but that their co-stimulatory activity may be detri-
of a clinical trial in stroke patients with the anti-ICAM-1 antibody mentally affected by stroke through mechanisms as of yet uncovered
enlimomab which, contrary to expectations, resulted in significant (Hug et al., 2011).
worsening of the clinical outcome (Enlimomab Acute Stroke Trial While leukocyte populations in the spleen have been shown to be
Investigators, 2001). This demonstrates the complex role of immune reduced 4 days after stroke, CD4+CD25+Foxp3+ T regulatory cells
cells in the development of stroke, and underscores that not only do (Tregs) and CD11b+ monocytes in the blood were highly increased at
they have pro-inflammatory functions potentially detrimental to recov- this time (Offner et al., 2006b), suggesting that splenic atrophy is not
ery, but also protective functions, particularly against stroke-related only dependent upon splenocyte apoptosis but is also partly due to the
infections (as discussed above), and indiscriminate ablation of such migration of cells out of the spleen and into the blood to then travel to
cells may not represent the best course of action. Additionally, a better un- the injured CNS. Indeed, CD11b+ monocytes can eventually infiltrate
derstanding of the temporal patterns of entry of each specific population the brain parenchyma at the site of infarct and further extend the sec-
may be useful in the implementation of the appropriate immuno- ondary damage to the CNS tissue. The drastic reduction in splenic
modulatory strategy. T and B cells must be a contributing factor to the immuno-suppression
In spite of the influx of peripheral immune cells into the brain there is observed following stroke, which has been linked to the increased sus-
little evidence of damaging autoimmunity after stroke. The presentation ceptibility to infections of stroke patients (Harms et al., 2008). The abnor-
of CNS antigens to T and B cells should induce the production of mal production of Tregs could also represent an immunosuppressing
autoantigens, and in fact in patients with a history of stroke, high num- factor in itself. Indeed, several studies now indicate that an excessive
bers of T-cells reactive to CNS antigen can be found in the circulation Treg presence may impede immuno-surveillance against tumor cells
(Bornstein et al., 2001). Offner and colleagues have shown that lack of and may suppress the ability of CD4+ effector T cells to, for example,
an adaptive immune response will reduce infarct size suggesting that eliminate parasites (Belkaid et al., 2002; Mackroth et al., 2011).
T and B-cells do contribute to lesion volume (Offner et al., 2009). Howev- More recently, the contribution of regulatory B cells in experimen-
er, the re-establishment of the BBB post stroke means that autoreactive tal stroke has been uncovered (Ren et al., 2011). Previous studies had
immune cells will have little or no opportunity to interact with CNS described the potent regulatory effects of B lymphocytes on inflam-
antigens during the recovery period. Interestingly, this has shown to be matory responses (LeBien and Tedder, 2008) and depletion of B
worsened in animal models using a peripheral inflammogen such as cells worsened disease severity in models of multiple sclerosis
LPS (Becker et al., 2005). (Matsushita et al., 2010). Ren and colleagues were the first to identify
Despite previous lateralization studies, work in both humans (Nilupul IL-10-secreting B regulatory cells as a major protective cell type in
Perera et al., 2006) and animal models (Dirnagl et al., 2007) has demon- stroke. Indeed, B cell deficiency exacerbated stroke outcomes and
strated that peripheral immune response post-ischemia is dependent dramatically increased inflammatory cell invasion into the brain,
not on anatomical location, but on infarct size. Work by Hug and col- suggesting that enhancement of regulatory B cells could have thera-
leagues have corroborated this finding, further showing that infarct vol- peutic applications in stroke (Ren et al., 2011).
ume directly correlates with post-stroke immune competence (Hug et Other aspects of the peripheral immune system may also be affected
al., 2011). Experimentally, this presents a problem, since both transient by ischemic events in the CNS. Offner's work demonstrating splenic atro-
and permanent MCAO models tend to produce large infarct volumes, af- phy also showed an increase in circulating monocytes (Offner et al.,
fecting both cortical and subcortical regions. Importantly, infarct size also 2006b). This has been suggested to be a ‘clean-up’ operation by the pe-
seems to be reflected by concentrations of CNS antigens myelin basic ripheral immune system (Manoonkitiwongsa et al., 2001), in order to re-
protein (MBP), creatine kinase-BB, neuron-specific enolase, S100beta, move necrotic tissue. However, considering the role of the macrophage
neurofilaments and portions of N-methyl-D-aspartate receptor in serum in the innate immune response to infection the increased susceptibility
samples from stroke patients (Bornstein et al., 2001; Dambinova et al., to infection post-stroke seems rather counter-intuitive. Other data
2003; Jauch et al., 2006). Lymphocytes from these patients show more show that the expression profile of genes on the surface of macrophages
reactivity against MBP than lymphocytes from multiple sclerosis patients and neutrophils is altered after stroke (Tang et al., 2006), suggesting the
(McQuillan et al., 2011; Mfonkeu et al., 2010). Since it is acknowledged possibility of immunomodulation, rather than immunodepression.
that an ischemic attack is a risk factor for developing dementia, this un-
derpins the hypothesis that autoimmune responses to the brain in stroke Conclusion
patients might contribute to the cognitive decline and progression of
white matter disease seen in some stroke patients (Pendlebury and The nature of the post-ischemic immune response is multifaceted
Rothwell, 2009). Thus, any attempt at immunomodulatory therapy in an- and complex, resulting in immunomodulation as well as immuno-
imals or humans, must be approached with caution given the paucity of depression. Factors such as current immune status and infarct volume
knowledge regarding the dysregulation of specific subsets of peripheral are capable of directly affecting the function of the immune system im-
immune cells after ischemic events of differing intensities. mediately after stroke and therefore, indirectly, affecting recovery and
32 R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33

repair from stroke. The window of opportunity for outside immuno- Dambinova, S.A., Khounteev, G.A., Izykenova, G.A., Zavolokov, I.G., Ilyukhina, A.Y.,
Skoromets, A.A., 2003. Blood test detecting autoantibodies to N-methyl-D-aspartate
modulatory therapy after stroke is relatively narrow and the current neuroreceptors for evaluation of patients with transient ischemic attack and stroke.
paucity of knowledge regarding the consequences of such therapies Clin. Chem. 49, 1752–1762.
means that they are often unwise. However, current work is aiming to del Zoppo, G.J., Sharp, F.R., Heiss, W.D., Albers, G.W., 2011. Heterogeneity in the
penumbra. J. Cereb. Blood Flow Metab. 31, 1836–1851.
further our knowledge of the immune system post-stroke and should Dirnagl, U., Klehmet, J., Braun, J.S., Harms, H., Meisel, C., Ziemssen, T., Prass, K., Meisel,
provide us with ample opportunity to improve clinical outcomes in A., 2007. Stroke-induced immunodepression: experimental evidence and clinical
the future. relevance. Stroke 38, 770–773.
Egea, J., Rosa, A.O., Sobrado, M., Gandia, L., Lopez, M.G., Garcia, A.G., 2007.
Neuroprotection afforded by nicotine against oxygen and glucose deprivation in
References hippocampal slices is lost in alpha7 nicotinic receptor knockout mice. Neuroscience
145, 866–872.
Ajmo Jr., C.T., Vernon, D.O., Collier, L., Hall, A.A., Garbuzova-Davis, S., Willing, A., Elenkov, I.J., Wilder, R.L., Chrousos, G.P., Vizi, E.S., 2000. The sympathetic nerve—an
Pennypacker, K.R., 2008. The spleen contributes to stroke-induced neurodegeneration. integrative interface between two supersystems: the brain and the immune
J. Neurosci. Res. 86, 2227–2234. system. Pharmacol. Rev. 52, 595–638.
Ajmo Jr., C.T., Collier, L.A., Leonardo, C.C., Hall, A.A., Green, S.M., Womble, T.A., Emsley, H.C., Smith, C.J., Tyrrell, P.J., Hopkins, S.J., 2008. Inflammation in acute ischemic
Cuevas, J., Willing, A.E., Pennypacker, K.R., 2009. Blockade of adrenoreceptors stroke and its relevance to stroke critical care. Neurocrit. Care 9, 125–138.
inhibits the splenic response to stroke. Exp. Neurol. 218, 47–55. Enlimomab Acute Stroke Trial Investigators, 2001. Use of anti-ICAM-1 therapy in ischemic
Anne, M., Juha, K., Timo, M., Mikko, T., Olli, V., Kyosti, S., Heikki, H., Vilho, M., 2007. stroke: results of the Enlimomab Acute Stroke Trial. Neurology 57, 1428–1434.
Neurohormonal activation in ischemic stroke: effects of acute phase distur- Fagan, S.C., Waller, J.L., Nichols, F.T., Edwards, D.J., Pettigrew, L.C., Clark, W.M., Hall, C.E.,
bances on long-term mortality. Curr. Neurovasc. Res. 4, 170–175. Switzer, J.A., Ergul, A., Hess, D.C., 2010. Minocycline to improve neurologic out-
Aslanyan, S., Weir, C.J., Diener, H.C., Kaste, M., Lees, K.R., 2004. Pneumonia and urinary come in stroke (MINOS): a dose-finding study. Stroke 41, 2283–2287.
tract infection after acute ischaemic stroke: a tertiary analysis of the GAIN Interna- Feinstein, D.L., Heneka, M.T., Gavrilyuk, V., Dello Russo, C., Weinberg, G., Galea, E., 2002.
tional trial. Eur. J. Neurol. 11, 49–53. Noradrenergic regulation of inflammatory gene expression in brain. Neurochem.
Astrup, J., Siesjo, B.K., Symon, L., 1981. Thresholds in cerebral ischemia — the ischemic Int. 41, 357–365.
penumbra. Stroke 12, 723–725. Franceschini, R., Gandolfo, C., Cataldi, A., Del Sette, M., Cianciosi, P., Finocchi, C., Rolandi,
Bao, Y., Kim, E., Bhosle, S., Mehta, H., Cho, S., 2010. A role for spleen monocytes in post- E., Barreca, T., 1994. Twenty-four-hour beta-endorphin secretory pattern in stroke
ischemic brain inflammation and injury. J. Neuroinflammation 7, 92. patients. Stroke 25, 2142–2145.
Becker, K.J., Kindrick, D.L., Lester, M.P., Shea, C., Ye, Z.C., 2005. Sensitization to brain Fujii, Y.X., Fujigaya, H., Moriwaki, Y., Misawa, H., Kasahara, T., Grando, S.A., Kawashima,
antigens after stroke is augmented by lipopolysaccharide. J. Cereb. Blood Flow K., 2007. Enhanced serum antigen-specific IgG1 and proinflammatory cytokine
Metab. 25, 1634–1644. production in nicotinic acetylcholine receptor alpha7 subunit gene knockout
Belkaid, Y., Piccirillo, C.A., Mendez, S., Shevach, E.M., Sacks, D.L., 2002. CD4+CD25+ mice. J. Neuroimmunol. 189, 69–74.
regulatory T cells control Leishmania major persistence and immunity. Nature Galea, E., Heneka, M.T., Dello Russo, C., Feinstein, D.L., 2003. Intrinsic regulation of brain
420, 502–507. inflammatory responses. Cell. Mol. Neurobiol. 23, 625–635.
Blond, D., Campbell, S.J., Butchart, A.G., Perry, V.H., Anthony, D.C., 2002. Differential in- Gelderblom, M., Leypoldt, F., Steinbach, K., Behrens, D., Choe, C.U., Siler, D.A.,
duction of interleukin-1beta and tumour necrosis factor-alpha may account for Arumugam, T.V., Orthey, E., Gerloff, C., Tolosa, E., Magnus, T., 2009. Temporal and
specific patterns of leukocyte recruitment in the brain. Brain Res. 958, 89–99. spatial dynamics of cerebral immune cell accumulation in stroke. Stroke 40,
Bornstein, N.M., Aronovich, B., Korczyn, A.D., Shavit, S., Michaelson, D.M., Chapman, J., 2001. 1849–1857.
Antibodies to brain antigens following stroke. Neurology 56, 529–530. Gendron, A., Teitelbaum, J., Cossette, C., Nuara, S., Dumont, M., Geadah, D., du Souich, P.,
Borovikova, L.V., Ivanova, S., Nardi, D., Zhang, M., Yang, H., Ombrellino, M., Tracey, K.J., Kouassi, E., 2002. Temporal effects of left versus right middle cerebral artery occlu-
2000. Role of vagus nerve signaling in CNI-1493-mediated suppression of acute sion on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain
inflammation. Auton. Neurosci. 85, 141–147. Res. 955, 85–97.
Bowes, M.P., Zivin, J.A., Rothlein, R., 1993. Monoclonal antibody to the ICAM-1 adhesion Haddad, J.J., Saade, N.E., Safieh-Garabedian, B., 2002. Cytokines and neuro-immune-
site reduces neurological damage in a rabbit cerebral embolism stroke model. Exp. endocrine interactions: a role for the hypothalamic-pituitary-adrenal revolving
Model. 119, 215–219. axis. J. Neuroimmunol. 133, 1–19.
Campbell, S.J., Deacon, R.M., Jiang, Y., Ferrari, C., Pitossi, F.J., Anthony, D.C., 2007. Haeusler, K.G., Schmidt, W.U., Fohring, F., Meisel, C., Helms, T., Jungehulsing, G.J., Nolte,
Overexpression of IL-1beta by adenoviral-mediated gene transfer in the rat brain C.H., Schmolke, K., Wegner, B., Meisel, A., Dirnagl, U., Villringer, A., Volk, H.D., 2008.
causes a prolonged hepatic chemokine response, axonal injury and the suppression Cellular immunodepression preceding infectious complications after acute ischemic
of spontaneous behaviour. Neurobiol. Dis. 27, 151–163. stroke in humans. Cerebrovasc. Dis. 25, 50–58.
Chamorro, A., Horcajada, J.P., Obach, V., Vargas, M., Revilla, M., Torres, F., Cervera, A., Hankey, G.J., 2006. Recurrent stroke rate was higher than coronary heart disease rate in
Planas, A.M., Mensa, J., 2005. The early systemic prophylaxis of infection after the first year after stroke but similar after the first year. ACP J. Club 144, 50.
stroke study: a randomized clinical trial. Stroke 36, 1495–1500. Hansson, G.K., Libby, P., 2006. The immune response in atherosclerosis: a double-edged
Chamorro, A., Urra, X., Planas, A.M., 2007. Infection after acute ischemic stroke: sword. Nat. Rev. Immunol. 6, 508–519.
a manifestation of brain-induced immunodepression. Stroke 38, 1097–1103. Harms, H., Prass, K., Meisel, C., Klehmet, J., Rogge, W., Drenckhahn, C., Gohler, J., Bereswill,
Chapman, K.Z., Dale, V.Q., Denes, A., Bennett, G., Rothwell, N.J., Allan, S.M., McColl, B.W., S., Gobel, U., Wernecke, K.D., Wolf, T., Arnold, G., Halle, E., Volk, H.D., Dirnagl, U.,
2009. A rapid and transient peripheral inflammatory response precedes brain in- Meisel, A., 2008. Preventive antibacterial therapy in acute ischemic stroke: a random-
flammation after experimental stroke. J. Cereb. Blood Flow Metab. 29, 1764–1768. ized controlled trial. PLoS One 3, e2158.
Chen, H., Chopp, M., Zhang, R.L., Bodzin, G., Chen, Q., Rusche, J.R., Todd III, R.F., 1994a. Hayakawa, K., Mishima, K., Nozako, M., Hazekawa, M., Mishima, S., Fujioka, M., Orito, K.,
Anti-CD11b monoclonal antibody reduces ischemic cell damage after transient Egashira, N., Iwasaki, K., Fujiwara, M., 2008. Delayed treatment with minocycline
focal cerebral ischemia in rat. Ann. Neurol. 35, 447–452. ameliorates neurologic impairment through activated microglia expressing a
Chen, H., Chopp, M., Zhang, R.L., Bodzin, G., Chen, Q., Rusche, J.R., Todd III, R.F., 1994b. high-mobility group box1-inhibiting mechanism. Stroke 39, 951–958.
Anti-CD11b monoclonal antibody reduces ischemic cell damage after transient Heijnen, C.J., 2007. Receptor regulation in neuroendocrine-immune communication:
focal cerebral ischemia in rat. Ann. Neurol. 35, 458–463. current knowledge and future perspectives. Brain Behav. Immun. 21, 1–8.
Chopp, M., Zhang, R.L., Chen, H., Li, Y., Jiang, N., Rusche, J.R., 1994. Post ischemic admin- Hewlett, K.A., Corbett, D., 2006. Delayed minocycline treatment reduces long-term
istration of an anti-Mac-1 antibody reduces ischemic cell damage after transient functional deficits and histological injury in a rodent model of focal ischemia. Neurosci-
middle cerebral artery occlusion in the rat. Stroke 25, 869–876. ence 141, 27–33.
Clark, W.M., Madden, K.P., Rothlein, R., Zivin, J.A., 1991a. Reduction of central nervous Hiramoto, T., Chida, Y., Sonoda, J., Yoshihara, K., Sudo, N., Kubo, C., 2008. The hepatic
system ischemic injury by monoclonal antibody to intercellular adhesion mole- vagus nerve attenuates Fas-induced apoptosis in the mouse liver via alpha7 nicotinic
cule. J. Neurosurg. 75, 623–627. acetylcholine receptor. Gastroenterology 134, 2122–2131.
Clark, W.M., Madden, K.P., Rothlein, R., Zivin, J.A., 1991b. Reduction of central nervous Hossmann, K.A., 2006. Pathophysiology and therapy of experimental stroke. Cell. Mol.
system ischemic injury in rabbits using leukocyte adhesion antibody treatment. Neurobiol. 26, 1057–1083.
Stroke 22, 877–883. Hug, A., Dalpke, A., Wieczorek, N., Giese, T., Lorenz, A., Auffarth, G., Liesz, A., Veltkamp,
Clausen, B.H., Lambertsen, K.L., Meldgaard, M., Finsen, B., 2005. A quantitative in situ R., 2009. Infarct volume is a major determiner of post-stroke immune cell function
hybridization and polymerase chain reaction study of microglial-macrophage ex- and susceptibility to infection. Stroke 40, 3226–3232.
pression of interleukin-1beta mRNA following permanent middle cerebral artery Hug, A., Liesz, A., Muerle, B., Zhou, W., Ehrenheim, J., Lorenz, A., Dalpke, A., Veltkamp, R.,
occlusion in mice. Neuroscience 132, 879–892. 2011. Reduced efficacy of circulating costimulatory cells after focal cerebral ischemia.
Colivicchi, F., Bassi, A., Santini, M., Caltagirone, C., 2004. Cardiac autonomic derangement and Stroke 42, 3580–3586.
arrhythmias in right-sided stroke with insular involvement. Stroke 35, 2094–2098. Iadecola, C., Anrather, J., 2011. The immunology of stroke: from mechanisms to trans-
Czech, B., Pfeilschifter, W., Mazaheri-Omrani, N., Strobel, M.A., Kahles, T., Neumann- lation. Nat. Med. 17, 796–808.
Haefelin, T., Rami, A., Huwiler, A., Pfeilschifter, J., 2009. The immunomodulatory Izci, Y., 2010. Splenectomy may be a prophylactic treatment for cerebral ischemia?
sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neuro- Med. Hypotheses 75, 347–349.
logical outcome in a mouse model of cerebral ischemia. Biochem. Biophys. Res. Jauch, E.C., Lindsell, C., Broderick, J., Fagan, S.C., Tilley, B.C., Levine, S.R., 2006. Association
Commun. 389, 251–256. of serial biochemical markers with acute ischemic stroke: the National Institute of
Czlonkowska, A., Cyrta, B., Korlak, J., 1979. Immunological observations on patients Neurological Disorders and Stroke recombinant tissue plasminogen activator Stroke
with acute cerebral vascular disease. J. Neurol. Sci. 43, 455–464. Study. Stroke 37, 2508–2513.
R. Brambilla et al. / Molecular and Cellular Neuroscience 53 (2013) 26–33 33

Jiang, N., Zhang, R.L., Chen, H., Chopp, M., 1994. Anti-CD11b monoclonal antibody Pendlebury, S.T., Rothwell, P.M., 2009. Prevalence, incidence, and factors associated with
reduces ischemic cell damage after transient (2h) but not after permanent MCA pre-stroke and post-stroke dementia: a systematic review and meta-analysis. Lancet
occlusion in the rat. Neurosci. Res. Commun. 15, 85–94. Neurol. 8, 1006–1018.
Jiang, N., Moyle, M., Soule, H.R., Rote, W.E., Chopp, M., 1995. Neutrophil inhibitory factor Prass, K., Meisel, C., Hoflich, C., Braun, J., Halle, E., Wolf, T., Ruscher, K., Victorov, I.V., Priller,
is neuroprotective after focal ischemia in rats. Ann. Neurol. 38 (6), 935–942. J., Dirnagl, U., Volk, H.D., Meisel, A., 2003. Stroke-induced immunodeficiency pro-
Johnston, K.C., Li, J.Y., Lyden, P.D., Hanson, S.K., Feasby, T.E., Adams, R.J., Faught Jr., R.E., motes spontaneous bacterial infections and is mediated by sympathetic activation
Haley Jr., E.C., 1998. Medical and neurological complications of ischemic stroke: reversal by poststroke T helper cell type 1-like immunostimulation. J. Exp. Med.
experience from the RANTTAS trial. RANTTAS Investigators. Stroke 29, 447–453. 198, 725–736.
Katzan, I.L., Cebul, R.D., Husak, S.H., Dawson, N.V., Baker, D.W., 2003. The effect of pneumo- Ren, X., Akiyoshi, K., Dziennis, S., Vandenbark, A.A., Herson, P.S., Hurn, P.D., Offner, H.,
nia on mortality among patients hospitalized for acute stroke. Neurology 60, 620–625. 2011. Regulatory B cells limit CNS inflammation and neurologic deficits in murine
Kohm, A.P., Sanders, V.M., 2001. Norepinephrine and beta 2-adrenergic receptor stimulation experimental stroke. J. Neurosci. 31, 8556–8563.
regulate CD4+ T and B lymphocyte function in vitro and in vivo. Pharmacol. Rev. 53, Rodriguez-Yanez, M., Davalos, A., Castillo, J., 2008. Withdrawal from statins: implications
487–525. for secondary stroke prevention and acute treatment. Int. J. Stroke 3, 85–87.
Krugers, H.J., Knollema, S., Kemper, R.H., Ter Horst, G.J., Korf, J., 1995. Down-regulation Ryan, K.J., Griffin, E.W., Connor, T.J., 2011. Complementary anti-inflammatory actions of
of the hypothalamo-pituitary-adrenal axis reduces brain damage and number of the beta-adrenoceptor agonist clenbuterol and the glucocorticoid dexamethasone
seizures following hypoxia/ischaemia in rats. Brain Res. 690, 41–47. in rat brain. J. Neuroimmunol. 232, 209–216.
Lambertsen, K.L., Biber, K., Finsen, B., 2012. Inflammatory cytokines in experimental Savitz, S.I., Erhardt, J.A., Anthony, J.V., Gupta, G., Li, X., Barone, F.C., Rosenbaum, D.M.,
and human stroke. J. Cereb. Blood Flow Metab. 32, 1677–1698. 2000. The novel beta-blocker, carvedilol, provides neuroprotection in transient
LeBien, T.W., Tedder, T.F., 2008. B lymphocytes: how they develop and function. Blood focal stroke. J. Cereb. Blood Flow Metab. 20, 1197–1204.
112, 1570–1580. Schulte-Herbruggen, O., Quarcoo, D., Meisel, A., Meisel, C., 2009. Differential affec-
Lipton, P., 1999. Ischemic cell death in brain neurons. Physiol. Rev. 79, 1431–1568. tion of intestinal immune cell populations after cerebral ischemia in mice.
Liu, Z., Fan, Y., Won, S.J., Neumann, M., Hu, D., Zhou, L., Weinstein, P.R., Liu, J., 2007. Neuroimmunomodulation 16, 213–218.
Chronic treatment with minocycline preserves adult new neurons and reduces Schwarz, S., Al-Shajlawi, F., Sick, C., Meairs, S., Hennerici, M.G., 2008. Effects of prophylactic
functional impairment after focal cerebral ischemia. Stroke 38, 146–152. antibiotic therapy with mezlocillin plus sulbactam on the incidence and height of fever
Mackroth, M.S., Malhotra, I., Mungai, P., Koech, D., Muchiri, E., King, C.L., 2011. Human after severe acute ischemic stroke: the Mannheim infection in stroke study (MISS).
cord blood CD4+CD25hi regulatory T cells suppress prenatally acquired T cell Stroke 39, 1220–1227.
responses to Plasmodium falciparum antigens. J. Immunol. 186, 2780–2791. Shimizu, J., Yamazaki, S., Sakaguchi, S., 1999. Induction of tumor immunity by removing
Maes, M., Kubera, M., Leunis, J.C., 2008. The gut-brain barrier in major depression: CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity.
intestinal mucosal dysfunction with an increased translocation of LPS from gram J. Immunol. 163, 5211–5218.
negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology Shytle, R.D., Mori, T., Townsend, K., Vendrame, M., Sun, N., Zeng, J., Ehrhart, J., Silver,
of depression. Neuro Endocrinol. Lett. 29, 117–124. A.A., Sanberg, P.R., Tan, J., 2004. Cholinergic modulation of microglial activation
Manoonkitiwongsa, P.S., Jackson-Friedman, C., McMillan, P.J., Schultz, R.L., Lyden, P.D., by alpha 7 nicotinic receptors. J. Neurochem. 89, 337–343.
2001. Angiogenesis after stroke is correlated with increased numbers of macrophages: Stevens, R.D., Nyquist, P.A., 2007. The systemic implications of aneurysmal subarachnoid
the clean-up hypothesis. J. Cereb. Blood Flow Metab. 21, 1223–1231. hemorrhage. J. Neurol. Sci. 261, 143–156.
Marrif, H., Juurlink, B.H., 1999. Astrocytes respond to hypoxia by increasing glycolytic Tang, Y., Xu, H., Du, X., Lit, L., Walker, W., Lu, A., Ran, R., Gregg, J.P., Reilly, M., Pancioli,
capacity. J. Neurosci. Res. 57, 255–260. A., Khoury, J.C., Sauerbeck, L.R., Carrozzella, J.A., Spilker, J., Clark, J., Wagner, K.R.,
Marsh, B.J., Williams-Karnesky, R.L., Stenzel-Poore, M.P., 2009. Toll-like receptor signaling Jauch, E.C., Chang, D.J., Verro, P., Broderick, J.P., Sharp, F.R., 2006. Gene expression
in endogenous neuroprotection and stroke. Neuroscience 158, 1007–1020. in blood changes rapidly in neutrophils and monocytes after ischemic stroke in
Marz, P., Cheng, J.G., Gadient, R.A., Patterson, P.H., Stoyan, T., Otten, U., Rose-John, S., humans: a microarray study. J. Cereb. Blood Flow Metab. 26, 1089–1102.
1998. Sympathetic neurons can produce and respond to interleukin 6. Proc. Natl. Tascilar, N., Irkorucu, O., Tascilar, O., Comert, F., Eroglu, O., Bahadir, B., Cakmak, G.K.,
Acad. Sci. U. S. A. 95, 3251–3256. Ankarali, H., Sayan, H., 2010. Bacterial translocation in experimental stroke: what
Matsushita, T., Horikawa, M., Iwata, Y., Tedder, T.F., 2010. Regulatory B cells (B10 cells) happens to the gut barrier? Bratisl. Lek. Listy 111, 194–199.
and regulatory T cells have independent roles in controlling experimental autoim- Tokgozoglu, S.L., Batur, M.K., Topuoglu, M.A., Saribas, O., Kes, S., Oto, A., 1999. Effects of stroke
mune encephalomyelitis initiation and late-phase immunopathogenesis. J. Immunol. localization on cardiac autonomic balance and sudden death. Stroke 30, 1307–1311.
185, 2240–2252. Turnbull, A.V., Rivier, C.L., 1999. Regulation of the hypothalamic-pituitary-adrenal axis
McColl, B.W., Allan, S.M., Rothwell, N.J., 2009. Systemic infection, inflammation and by cytokines: actions and mechanisms of action. Physiol. Rev. 79, 1–71.
acute ischemic stroke. Neuroscience 158, 1049–1061. Uehara, A., Gottschall, P.E., Dahl, R.R., Arimura, A., 1987. Interleukin-1 stimulates ACTH
McQuillan, J.A., Mitchell, A.J., Ho, Y.F., Combes, V., Ball, H.J., Golenser, J., Grau, G.E., Hunt, release by an indirect action which requires endogenous corticotropin releasing
N.H., 2011. Coincident parasite and CD8 T cell sequestration is required for devel- factor. Endocrinology 121, 1580–1582.
opment of experimental cerebral malaria. Int. J. Parasitol. 41, 155–163. Vendrame, M., Cassady, J., Newcomb, J., Butler, T., Pennypacker, K.R., Zigova, T.,
Mergenthaler, P., Dirnagl, U., Meisel, A., 2004. Pathophysiology of stroke: lessons from Sanberg, C.D., Sanberg, P.R., Willing, A.E., 2004. Infusion of human umbilical cord
animal models. Metab. Brain Dis. 19, 151–167. blood cells in a rat model of stroke dose-dependently rescues behavioral deficits
Mfonkeu, J.B., Gouado, I., Kuate, H.F., Zambou, O., Combes, V., Grau, G.E., Zollo, P.H., and reduces infarct volume. Stroke 35, 2390–2395.
2010. Biochemical markers of nutritional status and childhood malaria severity Vermeij, F.H., Scholte op Reimer, W.J., de Man, P., van Oostenbrugge, R.J., Franke, C.L., de
in Cameroon. Br. J. Nutr. 104, 886–892. Jong, G., de Kort, P.L., Dippel, D.W., 2009. Stroke-associated infection is an independent
Mirabelli-Badenier, M., Braunersreuther, V., Viviani, G.L., Dallegri, F., Quercioli, A., risk factor for poor outcome after acute ischemic stroke: data from the Netherlands
Veneselli, E., Mach, F., Montecucco, F., 2011. CC and CXC chemokines are pivotal Stroke Survey. Cerebrovasc. Dis. 27, 465–471.
mediators of cerebral injury in ischaemic stroke. Thromb. Haemost. 105, 409–420. Vogelgesang, A., May, V.E., Grunwald, U., Bakkeboe, M., Langner, S., Wallaschofski, H.,
Newberry, R.D., 2008. Intestinal lymphoid tissues: is variety an asset or a liability? Curr. Kessler, C., Broker, B.M., Dressel, A., 2010. Functional status of peripheral blood
Opin. Gastroenterol. 24, 121–128. T-cells in ischemic stroke patients. PLoS One 5, e8718.
Newcomb, J.D., Ajmo Jr., C.T., Sanberg, C.D., Sanberg, P.R., Pennypacker, K.R., Willing, Westendorp, W.F., Nederkoorn, P.J., Vermeij, J.D., Dijkgraaf, M.G., van de Beek, D., 2011.
A.E., 2006. Timing of cord blood treatment after experimental stroke determines Post-stroke infection: a systematic review and meta-analysis. BMC Neurol. 11, 110.
therapeutic efficacy. Cell Transplant. 15, 213–223. Williams, J.B., Jauch, E.C., Lindsell, C.J., Campos, B., 2007. Endothelial microparticle
Nilupul Perera, M., Ma, H.K., Arakawa, S., Howells, D.W., Markus, R., Rowe, C.C., Donnan, levels are similar in acute ischemic stroke and stroke mimics due to activation
G.A., 2006. Inflammation following stroke. J. Clin. Neurosci. 13, 1–8. and not apoptosis/necrosis. Acad. Emerg. Med. 14, 685–690.
Offner, H., Subramanian, S., Parker, S.M., Afentoulis, M.E., Vandenbark, A.A., Hurn, P.D., Woiciechowsky, C., Asadullah, K., Nestler, D., Eberhardt, B., Platzer, C., Schoning, B.,
2006a. Experimental stroke induces massive, rapid activation of the peripheral Glockner, F., Lanksch, W.R., Volk, H.D., Docke, W.D., 1998. Sympathetic activation
immune system. J. Cereb. Blood Flow Metab. 26, 654–665. triggers systemic interleukin-10 release in immunodepression induced by brain
Offner, H., Subramanian, S., Parker, S.M., Wang, C., Afentoulis, M.E., Lewis, A., Vandenbark, injury. Nat. Med. 4, 808–813.
A.A., Hurn, P.D., 2006b. Splenic atrophy in experimental stroke is accompanied by in- Woiciechowsky, C., Schoning, B., Lanksch, W.R., Volk, H.D., Docke, W.D., 1999.
creased regulatory T cells and circulating macrophages. J. Immunol. 176, 6523–6531. Mechanisms of brain-mediated systemic anti-inflammatory syndrome causing
Offner, H., Vandenbark, A.A., Hurn, P.D., 2009. Effect of experimental stroke on peripheral immunodepression. J. Mol. Med. (Berl.) 77, 769–780.
immunity: CNS ischemia induces profound immunosuppression. Neuroscience 158, Wong, C.H., Jenne, C.N., Lee, W.Y., Leger, C., Kubes, P., 2011. Functional innervation of
1098–1111. hepatic iNKT cells is immunosuppressive following stroke. Science 334, 101–105.
Oto, J., Suzue, A., Inui, D., Fukuta, Y., Hosotsubo, K., Torii, M., Nagahiro, S., Nishimura, M., Young, J.B., Fish, S., Landsberg, L., 1983. Sympathetic nervous system and adrenal medul-
2008. Plasma proinflammatory and anti-inflammatory cytokine and catecholamine lary responses to ischemic injury in mice. Am. J. Physiol. 245, E67–E73.
concentrations as predictors of neurological outcome in acute stroke patients. J. Anesth. Zemke, D., Majid, A., 2004. The potential of minocycline for neuroprotection in human
22, 207–212. neurologic disease. Clin. Neuropharmacol. 27, 293–298.
Ottani, A., Giuliani, D., Mioni, C., Galantucci, M., Minutoli, L., Bitto, A., Altavilla, D., Zaffe, D., Zhang, R.L., Chopp, M., Jiang, N., Tang, W.X., Prostak, J., Manning, A.M., 1995a. Anti-
Botticelli, A.R., Squadrito, F., Guarini, S., 2009. Vagus nerve mediates the protective ICAM-1 antibody reduces ischemic cell damage after transient but not permanent
effects of melanocortins against cerebral and systemic damage after ischemic stroke. MCA occlusion in the Wistar rat. Stroke 26, 1438–1443.
J. Cereb. Blood Flow Metab. 29, 512–523. Zhang, Z.G., Chopp, M., Tang, W.X., Jiang, N., Zhang, R.L., 1995b. Post-ischemic treat-
Pankoui Mfonkeu, J.B., Gouado, I., Fotso Kuate, H., Zambou, O., Amvam Zollo, P.H., Grau, ment (24h) with anti-CDllb and anti-CD18 monoclonal antibodies are neuro-
G.E., Combes, V., 2010. Elevated cell-specific microparticles are a biological marker protective after transient (2h) focal cerebral ischemia in the rat. Brain Res. 698,
for cerebral dysfunctions in human severe malaria. PLoS One 5, e13415. 79–85.

You might also like