• No results found

Inflammation and epilepsy: the contribution of astrocytes - 1.2: Astrocytes immune response in epilepsy

N/A
N/A
Protected

Academic year: 2021

Share "Inflammation and epilepsy: the contribution of astrocytes - 1.2: Astrocytes immune response in epilepsy"

Copied!
27
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE (Digital Academic Repository)

Inflammation and epilepsy: the contribution of astrocytes

Zurolo, E.

Publication date

2013

Link to publication

Citation for published version (APA):

Zurolo, E. (2013). Inflammation and epilepsy: the contribution of astrocytes.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

1.2 Astrocyte immune responses in epilepsy

Eleonora Aronica

1,2

Teresa Ravizza

3

, Emanuele Zurolo

1

,

Annamaria Vezzani

3

1Department (Neuro) Pathology, Academisch Medisch Centrum, Amsterdam, and 2 Epilepsy Institute in The Netherlands Foundation

(Stichting Epilepsie Instellingen Nederland, SEIN), Heemstede, The Netherlands.

3Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156 Milano; Italy

Glia. 2012 Aug;60(8):1258-68.

Abstract

Astrocytes, the major glial cell type of the central nervous system (CNS), are known to play a major role in the regulation of the immune/inflammatory response in several human CNS diseases. In epilepsy-associated pathologies, the presence of astrogliosis has stimulated ex-tensive research focused on the role of reactive astrocytes in the pathophysiological pro-cesses that underlie the development of epilepsy. In brain tissue from patients with epi-lepsy, astrocytes undergo significant changes in their physiological properties, including the activation of inflammatory pathways. Accumulating experimental evidence suggests that proinflammatory molecules can alter glio-neuronal communications contributing to the generation of seizures and seizure-related neuronal damage. In particular, both in vitro and

in vivo data point to the role of astrocytes as both major source and target of epileptogenic

inflammatory signaling. In this context, understanding the astroglial inflammatory response occurring in epileptic brain tissue may provide new strategies for targeting astrocyte-medi-ated epileptogenesis.

This article reviews current evidence regarding the role of astrocytes in the regulation of the innate immune responses in epilepsy. Both clinical observations in drug-resistant human ep-ilepsies and experimental findings in clinically relevant models will be discussed and elabo-rated, highlighting specific inflammatory pathways (such as interleukin-1β/toll-like receptor 4) that could be potential targets for antiepileptic, disease-modifying therapeutic strategies.

(3)

Astrocytes as key players in brain inflammation

The central nervous system (CNS) has commonly been considered an immune-privileged site in the presence of an intact blood brain barrier (BBB). However, this concept is gradually changing as a result of recent developments in the field of innate immunity supporting the role of CNS-resident cells acting as innate-immune-competent cells; [for reviews see (Amor et al. 2010; McNaull et al. 2010; Schwartz and Kipnis 2011)]. Both innate and adaptive in-flammatory responses occur in the CNS. The innate immune response is a non-specific, acute defense against external agents or local injuries, and cell types chiefly involved in this response include monocytes/macrophages and microglia. The adaptive immune response is antigen-specific,and B and T lymphocytes are the key players in adaptive immunity. Com-munication between the innate and adaptive responses involves cell-cell interactions, as well as soluble factors such as cytokines and chemokines. In earlier studies, a considerable amount of attention was focused on the immune function of microglial cells, which are gen-erally considered to be the “CNS-based resident macrophages” (Aloisi 2001; Graeber and Streit 1990). However, over the past decade there has been accumulating evidence support-ing the central role of astrocytes in the CNS innate immune response induced by a variety of insults. Astrocytes have been shown to initiate, regulate and amplify immune-mediated mechanisms involved in different human CNS diseases, including epilepsy [for reviews see (Farina et al. 2007; Seifert et al. 2010)]. In this review we will largely focus on protoplas-mic astrocytes that predominate in gray matter, and fibrous astrocytes that predominate in white matter. Astrocytes undergo complex morphological and functional changes. These changes may vary with the severity and the type of insult and a continuum of progressive morphological alterations can be observed in human brain tissue under different pathologi-cal conditions [for review see (Sofroniew and Vinters 2010)].

Astrocytes as innate-immune-competent cells

Astrocytes as antigen-presenting cells

In contrast to the well-established role of microglia as antigen presenting cells (APCs) the role of astrocytes in antigen presentation is still a matter of debate. Previous studies have shown the induction of major histocompatibility (MHC) class I and II molecules on interferon-γ (IFN-γ) exposure in vitro [for reviews see (Aloisi et al. 2000a; Dong and Benveniste 2001)]. However, there are discrepancies in the literature concerning the ability of astrocytes to act as fully competent APCs (Aloisi et al. 2000a; Becher et al. 2000; Cornet et al. 2000; Cross and Ku 2000; Dong and Benveniste 2001; Girvin et al. 2002; Weber et al. 1994). Moreover, the demonstration of MHC-positive astrocytes in pathological human brain remains a contro-versial topic, even in inflammatory neurological disorders such as multiple sclerosis (MS) (De

(4)

Keyser et al. 2003; Gobin et al. 2001; Traugott and Raine 1985). In Rasmussen’s encephalitis (RE), a severe inflammatory epileptic encephalopathy of childhood, expression of MHC class I molecules has been reported in astrocytes (Bauer et al. 2007; Farrell et al. 1995), suggest-ing an MHC class I–restricted T-cell response as a possible mechanism for the occurrence of the astrocytic degeneration observed in RE (Bauer et al. 2007). However, the final effect of astrocyte-T-lymphocyte interactions is complex and depends on the type of responding T cells (Th1 or Th2 cells). Accordingly, whereas microglia may activate both Th1 and Th2 cells, astrocytes have been shown to stimulate mainly Th2 responses, providing homeostat-ic mechanisms whhomeostat-ich may limit brain inflammation (Aloisi et al. 1998; Aloisi et al. 2000b). The physiological role of MHC molecules (MHC class I), as well as of other immunologi-cal molecules/receptors expressed by astrocytes during brain development, is another im-portant area for future research. Recent studies suggest that immune molecules critically modulate the development and function of the CNS [for review see (Glynn et al. 2011)].

Astrocytes as source and target of inflammatory molecules

Astrocytes represent an important source of immunologically relevant cytokines and chemokines. In vitro studies document the ability of astrocytes (particularly reactive astro-cytes) to produce cytokines such as interleukin(IL) -1β, IL-6, tumor necrosis factor (TNF)-α,

transforming growth factor beta (TGF)-β and chemokines, such as monocyte chemoattract-ant protein-1 (MCP-1; chemokine, C-C motif, ligand 2; CCL2), which are highly expressed in both experimental and human epileptogenic brain tissue [for reviews see (Aronica and Crino 2011; Vezzani et al. 2008b) Fig. 1]. The expression pattern and the role of astrocyte-derived inflammatory molecules in seizure generation and progression will be discussed in the fol-lowing sections.

Astrocytes are also the target of inflammatory molecules which, through the activation of specific receptors (including pattern-recognition receptors (PRRs) and related intracellular signaling pathways), may aggravate astrogliosis and amplify the pro-epileptogenic inflam-matory signaling [for reviews see (Aronica and Crino 2011; Farina et al. 2007; Sofroniew and Vinters 2010); Fig. 1].

Particular attention has recently been focused on the role of the IL-1 receptor/toll-like re-ceptor superfamily (IL-1R/TLR) in epilepsy (Aronica and Crino 2011; Maroso et al. 2011; Vezzani et al. 2010; Vezzani et al. 2011d; Vezzani et al. 2008b). The IL-1R/TLR superfamily comprises cell surface PRRs sharing a conserved region termed the Toll/IL-1R (TIR) domain (O’Neill 2008; O’Neill and Dinarello 2000). Several TLRs are expressed in human astrocytes

in vitro, including TLR2, TLR3 and TLR4 (Farina et al. 2007). However, whereas TLR3 shows

(5)

astrocytes have produced conflicting results, which may reflect differences in cell source and culture conditions (Crack and Bray 2007; Farina et al. 2007; Kielian 2006). Expression of IL-1R1 or TLRs in astrocytes has also been demonstrated in human brain, with low levels in resting astrocytes and upregulation in reactive astrocytes under different pathological conditions, including epilepsy [(Maroso et al. 2010; Maroso et al. 2011; Ravizza et al. 2006a; Ravizza et al. 2008a; Vezzani et al. 2011d; Zurolo et al. 2011); Fig.1].

TLRs have a key role in pathogen recognition (Kawai and Akira 2007), but in the absence of pathogens, TLR can be activated by endogenous molecules, named damage-associated molecular patterns (DAMPs), released from injured or activated cells. One of these mol-ecules is the high mobility group box 1 (HMGB1) (Bianchi and Manfredi 2009), a ubiquitous chromatin component that can be actively secreted by immuno-competent cells in response to immune challenges (Muller et al. 2004). Both in vitro and in vivo findings suggest that astrocytes are a source of extracellular HMGB1 (Maroso et al. 2010 ; Zurolo et al. 2011). In particular, HMGB1 release has been shown to be induced in both rat (Hayakawa et al. 2010) and human astrocytes in culture (Zurolo et al. 2011) in response to the pro-inflammatory Figure 1. Astrocytes as key players in neuroinflammation in the epileptic brain. Schematic represen-tation of the hypothetical cascade of inflammatory processes in which astrocytes are involved. Knowl-edge of the astrocyte immune-inflammatory function in epilepsy may create the basis for developing effective therapeutic strategies to control seizures.

(6)

cytokine IL-1β, and nuclear to cytoplasmic translocation has been observed in human and experimental epileptic tissue (Maroso et al. 2010). In addition, astrocytes have also been shown to respond to HMGB1 stimulation with induction of several inflammatory mediators (Pedrazzi et al. 2007).

Recently, a new class of regulators of the immune responses has been recognised in the form of microRNAs (miRNA), acting as post-transcriptional regulators of gene expression (Gantier 2010; Quinn and O’Neill 2011; Sonkoly et al. 2008). In particular, the miRNA-146a has been specifically associated with the regulation of TLR signaling (Cui et al. 2010; Quinn and O’Neill 2011; Sheedy and O’Neill 2008; Taganov et al. 2006). miRNA-146a is expressed in human brain, and astrocytes have been shown to be key players in the regulation of this miRNA in response to inflammatory molecules, such as IL-1β (Aronica et al. 2010; Cui et al. 2010).

Another important component of the innate immune response is the complement system; this consists of a variety of soluble and surface proteins which, when activated, result in a complex cascade of processes contributing to the amplification of the inflammatory re-sponse [for review see (Bonifati and Kishore 2007)]. Reactive astrocytes are a source of complement components and also express complement-regulatory proteins, as well as complement receptors [for review see (Farina et al. 2007); Fig. 1]. Complement activation products, such as C3, regulate cytokine synthesis, and cytokines (such as IL-1β) may also in-duce complement factor expression in human astrocytes (Barnum and Jones 1995; Bonifati and Kishore 2007; Veerhuis et al. 1999). Astrocytes can also contribute to regulation of this inflammatory pathway by induction of inhibitory factors, such as the complement factor H (CFH) (Aronica et al. 2007; Boon et al. 2009; Griffiths et al. 2009). In addition, recent evi-dence suggests an extensive and complex cross-regulation between complement and the TLRs, which deserves further investigation in astrocytes (Hajishengallis and Lambris 2010).

Astrocyte immune-inflammatory function and neurotrasmitter receptors

Signaling via neurotransmitter receptors provides an additional mechanism by which astro-cytes can sense and respond to changes in the extracellular environment, influencing the inflammatory and immune response under pathological conditions associated with astro-gliosis. An example is provided by the activation of astroglial G protein-coupled glutamate and purinergic receptors, the expression of which is deregulated in epileptogenic brain tis-sue (Byrnes et al. 2009; D’Antoni et al. 2008; Gomes et al. 2011; Hasko et al. 2005; Matute and Cavaliere 2011).

Both in vitro and in vivo studies suggest an up-regulation of group I and II metabotropic glu-tamate receptor (mGluR) subtypes (mGluR5 and mGluR3) in reactive astrocytes [(Aronica et

(7)

al. 2003; Aronica et al. 2005a,b; Aronica et al. 2000); for review see (D’Antoni et al. 2008)]. Activation of mGluR3 in human astrocytes in culture modulates the release of IL-6 in the presence of IL-1β, supporting the role of this receptor subtype in regulating the capacity of activated astrocytes to produce inflammatory cytokines (Aronica et al. 2005a).

Increasing evidence points towards a critical role of purinergic receptors in neuron–glia communication and neuroinflammation (Boison 2010; Gomes et al. 2011). Stimulation of the adenosine receptor (P1 receptor) A2B induces the release of IL-6 from astrocytes and the activation of the A3 receptor induces the synthesis of the chemokine MCP-1 [for reviews see (Abbracchio and Ceruti 2007; Gomes et al. 2011; Hasko et al. 2005)]. The P2 purinergic receptors modulate the cytokine-mediated signal transduction in human astrocytes in cul-ture (Liu et al. 2000). Although the expression of both P2X (4,6,7) and P2Y(1,2) receptor-subtypes has been reported in cultured astrocytes, P2Y rather than P2X receptor-receptor-subtypes have been suggested as being involved in the modulation of intracellular Ca2+ (Fischer et al.

2009). In human fetal astrocytes, the blockade of P2Y receptors affects both IL-1β and TNFa signaling (Liu et al. 2000), whereas the P2X7 receptor has been implicated in the regulation of chemokine synthesis in astrocytes (Panenka et al. 2001). In addition, inflammatory mol-ecules, such as IL-1β, may modulate the expression of adenosine kinase (ADK), providing a potential modulatory crosstalk between the astrocyte-based adenosine cycle and inflamma-tion (Aronica et al. 2011).

Finally, it has also been suggested that cannabinoid (CB) receptors, as mediators of endocan-nabinoid signaling, exert an immunomodulatory function on astrocytes (Sheng et al. 2005).

Astroglial inflammatory response in epilepsy

Increasing evidence supports the concept of activation of innate immune responses in both experimental and human epilepsy and the critical involvement of inflammatory processes in the etiopathogenesis of seizures (Aronica and Crino 2011; Vezzani et al. 2010; Vezzani et al. 2011d). In particular, recent clinical-neuropathological and experimental observations support the notion that dysregulation of the astrocyte immune-inflammatory function (dis-cussed above) is a common factor, which may predispose or directly contribute to the gen-eration of seizures and to seizure-related neuronal damage in epilepsy of various etiologies (Fig. 1). Current knowledge concerning common inflammatory signaling pathways involving astrocytes that may alter neuronal excitability is discussed below.

(8)

Astrocytes and inflammatory processes in patients with

medically refractory epilepsy

Reactive astrogliosis is a pathological hallmark of various types of medically refractory focal epilepsy, including epilepsy that develops following ischemic, traumatic, or infectious brain injury (Sofroniew and Vinters 2010). Reactive astrogliosis is also the pathological hallmark of two major epilepsy-associated pathologies [hippocampal sclerosis and focal malformations of cortical development (MCD; FCD and cortical tubers in tuberous sclerosis complexTSC)]. Hippocampal sclerosis (HS) is the most common neuropathological finding in patients un-dergoing surgery for intractable temporal lobe epilepsy [TLE; (Wieser 2004)]. Although specimens obtained from patients undergoing surgery for intractable TLE often represent the end-stage of the pathological cascade that leads to HS, histopathological and molecular analysis of this tissue is essential to confirm the relevance and cellular sources of inflamma-tory molecules and related signaling. Large-scale analysis of gene expression profiles sug-gests a prominent upregulation of genes related to astroglial activation and innate immune/ inflammatory response in human TLE [for review see (Aronica and Gorter 2007 )] This evi-dence, at gene expression level, has been confirmed by histopathological studies demon-strating the association between activated astrocytes and microglial cells and the induction of major proinflammatory pathways in human TLE (Aronica and Gorter 2007; Ravizza et al. 2008a; Vezzani et al. 2010).

The transcription factor nuclear factor-kappa B (NFkB) plays a central role in regulating im-mune and inflammatory responses, including the IL-1R/TLR signaling pathways (Oecking-haus et al. 2011). Activation of IL-1R1-mediated signaling in cells targeted by the released IL-1β induces, via an NFkB-dependent mechanism, the transcription of other genes encod-ing downstream mediators of inflammation, includencod-ing IL-6, TNF-α, cyclooxygenase-2 (Cox-2) or CCL2 (i.e. monocyte chemotactic protein-1) (Andjelkovic et al. 2000; Dinarello 2004; Meeuwsen et al. 2003). Crespel et al. (Crespel et al. 2002) reported NFkB over-expression in reactive astrocytes in human HS specimens. The activation of the NFkB signaling pathway in astrocytes has been confirmed by subsequent studies demonstrating prominent expres-sion in astrocytes of both L-1β and its functional receptor, IL-1R1, providing evidence of a persistent activation of this specific inflammatory pathway in human tissue from people with chronic epilepsy (Ravizza et al. 2008a). In addition, up-regulation of astroglial Cox-2 and CCL2 has been reported in human TLE (Desjardins et al. 2003; Holtman et al. 2009; Wu et al. 2008) suggesting the activation of a complex, highly interconnected, cytokine network. Human studies in TLE also support the involvement of the complement system. Expression of various complement components, such as C1q, C3c and C3d, has been observed in reac-tive astrocytes within the sclerotic hippocampus of people with TLE (Aronica et al. 2007).

(9)

Complement activation in astrocytes may regulate cytokine synthesis thus critically contrib-uting to the propagation and persistence of the inflammatory response.

The activation of the plasminogen system in astrocytes in human TLE may contribute to the regulation of the immune responses and related inflammation within the epileptic lesion (Benarroch 2007). Accordingly, in addition to neurons and microglial cells, expression of both tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) has been observed in reactive astrocytes (Iyer et al. 2010). Although PAs may contribute to the activa-tion of astrocytes (Gravanis and Tsirka 2005), astrocytes are also involved in the neutraliza-tion of tPA toxicity via an endocytic tPA receptor (Fernandez-Monreal et al. 2004).

Induction of the pathways discussed above (IL-1β, complement and PA) in perivascular as-trocytes suggests that the alterations of BBB permeability described in human TLE (Rigau et al. 2007; van Vliet et al. 2007) probably result from the convergence of the actions of dif-ferent inflammatory molecules released from parenchymal brain cells, then acting on blood vessels. In this context, the vascular endothelial growth factor A (VEGFA)-signaling pathway may also affect the integrity of the BBB (Schoch et al. 2002). VEGFA is upregulated in reac-tive astrocytes (including perivascular astrocytes) in human epileptogenic tissue [HS and FCD; (Boer et al. 2008; Morin-Brureau et al. 2011; Rigau et al. 2007)], and increased VEGFA expression has recently been reported within cortical tubers of people with TSC (Parker et al. 2011).

As mentioned above, the activation of the TLR signaling pathway in astrocytes plays a key role in the regulation of the innate immune response. Interestingly overexpression of TLR4 and its endogenous ligand HMGB1 has been demonstrated in human TLE in reactive as-trocyes, confirming the role of these cells as both source and target of HMGB1 (Maroso et al. 2010). The functional consequences of the astrocyte-mediated HMGB1-TLR signaling in ictogenesis and epileptogenesis in experimental models are discussed below (see Fig. 2). Reactive astrogliosis is also a major feature of focal MCD, such as FCD and cortical tubers in TSC (Blumcke et al. 2011; Blümcke et al. 2009; Orlova and Crino ; Sosunov et al. 2008; Wong 2008).

The association between astrogliosis and activation of inflammatory pathways is supported by gene expression analysis of cortical tubers in TSC; high expression levels have been ob-served for genes encoding complement components, chemokines, MHC elements and com-ponents of the IL-1R/TLR signaling pathways (Boer et al. 2010).

Activation of various proinflammatory molecules and related pathways has been confirmed at the cellular level in both TSC and FCD specimens, pointing to the central role of reactive astrocytes in the immune/inflammatory response of these developmental epileptogenic le-sions as well [for review see (Aronica and Crino 2011)].

(10)

A recent study (Zurolo et al. 2011) demonstrates the activation of the TLR signaling pathway in reactive astrocyes both in FCD type and TSC specimens, as shown by overexpression of TLR4 and RAGE (receptor for advanced glycation end products) and cytoplasmic transloca-tion of HMGB1 [as reported in human HS specimens; (Maroso et al. 2010)].

The transforming growth factor (TGF)-β mediated pathway is also upregulated in focal MCD (Boer et al. 2010; Kim et al. 2003). TGF-β is a multifunctional cytokine which, acting through its specific receptors, modulates the astrocyte immune responses and which has recently been suggested as playing a critical role in neocortical epileptogenesis acting through its specific receptors (Ivens et al. 2007).

A controversial issue is whether the dysregulation of astrocyte immune-inflammatory re-sponses is related to the underlying cellular pathology in epileptic tissue or whether it oc-curs as a consequence of recurrent seizures. The observation of prenatal TLRs and HMGB1 expression in giant cells within the tuber in TSC (Aronica et al. 2008; Samadani et al. 2007; Zurolo et al. 2011) suggests that the induction of these signaling pathways could be intrinsic to the developmental lesion per se, and that the development of seizures later in life could contribute to perpetuation of their activation.

These observations provide evidence of the potential role of astrocytes in the chronic in-Figure 2. Functional consequences of the production of inflammatory mediators by astrocytes.

Proepileptogenic brain injuries trigger brain inflammation, involving the induction/overexpression of cytokines (e.g. IL-1β), danger signals (e.g. HMGB1) and TLRs (e.g. TLR4) in parenchymal/perivascular astrocytes. This event leads to changes in brain physiology such as neuronal hyperexcitability, BBB dys-function and cell damage that can contribute to lower seizure threshold and trigger epileptogenesis.

(11)

flammatory state observed in focal MCD. Whether the dysregulation of astrocyte immune-inflammatory responses could contribute to progressive cognitive dysfunction in children deserves further investigation (Chew et al. 2006; Cohly and Panja 2005).

Finally, both gene expression and immunocytochemical studies suggest prominent activa-tion of major proinflammatory pathways within the astroglial component of highly epilepto-genic tumors (glioneuronal tumors, such as gangliogliomas) (Aronica et al. 2008; Samadani et al. 2007; Zurolo et al. 2011).

Astrocyte and brain inflammation in experimental models

of seizures and epilepsy

Experimental studies provided the first evidence showing a significant contribution of reac-tive astrocytes to the inflammatory processes developing after seizures or induced by an ep-ileptogenic brain injury. Acute seizures induced in rodents (following intracerebral applica-tion of kainate or bicuculline, or electrically induced status epilepticus) were shown rapidly to upregulate prototypical inflammatory cytokines in microglia and astrocytes in the brain areas where seizures originate and spread; as a consequence of this event, a downstream cascade of inflammatory mediators is transcriptionally upregulated in brain tissue similar to what has been shown in human epilepsy [(De Simoni et al. 2000; Gorter et al. 2006; Vezzani et al. 1999; Vezzani et al. 2000); reviewed by (Kulkarni and Dhir 2009; Vezzani et al. 2008a; Vezzani et al. 2011b)]. Activation of astrocytes in the absence of neuronal degeneration has been reported in a kindling model (Khurgel et al. 1995) and induction of GFAP has been observed even after a single electroconvulsive seizure (Steward et al. 1992). A detailed time-course analysis of these inflammatory processes occurring after status epilepticus in rats was instrumental in demonstrating that the mRNA of inflammatory mediators are induced within 30 minutes of seizure onset (De Simoni et al. 2000). The immunohistochemical analy-sis of IL-1β in status epilepticus models showed that the expression of this cytokine in micro-glia is time-locked to the occurrence of seizures and the extent of expression depends on the recurrence of seizures, while astrocytes appear to be involved in perpetuating inflammation even in the long-term after the initial injury (Ravizza et al., 2008a). Moreover, as in human epileptic tissue, astrocytes often express both the inflammatory mediator and the cognate cell signaling receptors, thus highlighting that these cells serve as sources and targets of in-flammatory molecules (reviewed in (Maroso et al. 2011; Vezzani et al. 2011c). Experimental findings clearly show that both parenchymal and perivascular astrocytes are activated and express inflammatory molecules in epilepsy models with functional consequences on BBB function (Fig. 2) (Bauer et al. 2008; Friedman et al. 2009; Ravizza et al. 2008a; van Vliet et al. 2009; Vezzani et al. 2011c). Notably, specific anti-inflammatory molecules, such as the IL-1

(12)

receptor antagonist (IL-1ra) (De Simoni et al. 2000 ) or the C59 inhibitor of the complement system (Aronica et al. 2007), are induced to a limited extent by seizures or following brain injury, suggesting that the mechanisms involved in the resolution of brain inflammation are not very efficient, and possibly explaining why inflammation is detrimental for tissue excit-ability and cell survival.

Although induction of various inflammatory molecules has been demonstrated in astrocytes in seizure models [reviewed in (Friedman and Dingledine 2011; Vezzani et al. 2008a; Weth-erington et al. 2008)], the IL-1/TLR is the first inflammatory signalling to be induced during innate immunity activation, either by a pathogen or a danger signal serving as endogenous ligand (Maroso et al. 2011; Vezzani et al. 2011c). IL-1/TLR signalling is rapidly induced by tis-sue injury or seizures in neurons, microglia and astrocytes resulting in transcriptional activa-tion of inflammatory genes, and other genes potentially involved in synaptic and molecular changes underlying epileptogenesis [reviewed in (Vezzani et al. 2011c)]. Two endogenous li-gands, IL-1β and HMGB1 (Fig. 2) arereleased by glial cells; IL-1β activates IL-1R1 and HMGB1 activates TLR4 in neurons with significant consequences for ictogenesis, mainly mediated by post-translational effects (see later).

Functional consequences of astrocyte-mediated brain

in-flammation on neuronal excitability

The activation of inflammatory pathways and the consequent release of inflammatory mol-ecules by astrocytes alter neural network excitability via induction of various mechanisms, with either direct or indirect impact on neuronal functions. Here we focus on the IL-1R1/ TLR4 signaling because of its prominent involvement in seizures and epileptogenesis [(Ma-roso et al. 2011; Vezzani et al. 2011c); Fig. 2].

IL-1β, by acting on IL-1R type 1, can inhibit the astrocytic reuptake of glutamate (Hu et al. 2000; Ye and Sontheimer 1996) and increases its glial release likely via induction of TNFa (Bezzi et al. 2001). These effects result in elevated extracellular glutamate levels, which in turn can promote tissue excitability. IL-1β can also increase neuronal glutamate release via the activation of inducible nitric oxide synthase in glial cells (Casamenti et al. 1999; Hewett et al. 1994). Astrocytic glutamate release may have a role in the genesis or strength of sei-zure-like events (Carmignoto and Fellin 2006; Tian et al. 2005).

In hippocampal neurons, IL-1R1 co-localizes with NMDA receptors, a subtype of glutamate receptor involved in the onset and spread of seizures. IL-1β potentiates NMDA receptor function in cultured hippocampal neurons (Lai et al. 2006; Viviani et al. 2003) by enhanc-ing N-Methyl-D-aspartate (NMDA)-mediated Ca2+ influx via IL-1R1 dependent activation of

(13)

mechanism (within minutes) involves ceramide-mediated activation of Src kinases (Viviani et al. 2003), and contributes to seizure generation and recurrence (Balosso, 2008). IL-1β also down-regulates AMPA receptor expression and phosphorylation in hippocampal neurons in a Ca2+- and NMDA-dependent manner (Lai et al. 2006).

Interactions of IL-1β with GABA-mediated inhibitory neurotransmission have also been re-ported. However, the results obtained are not consistent. Thus IL-1β can either decrease or increase GABA inhibition depending on the brain area (Alam, 2004; Wilkinson, 1993), the cytokine concentration (Wang, 2000; Zeise, 1997; Serantes, 2006) and the functional properties of the cells (Hori, 1988), highlighting a dual role of IL-1β in affecting GABAergic inhibitory system.

In the hippocampus, IL-1β affects synaptic transmission, and inhibits long-term potentiation

(LTP) via activation of Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) (Bellinger et al. 1993; O’Donnell et al. 2000; Schneider et al. 1998). This cytokine can also modulate neurotransmitter release via inhibition of voltage-dependent Ca2+ channels,

an effect that involves pertussis-sensitive G proteins and protein kinase C ( PKC) (Plata-Salaman and ffrench-Mullen 1994).

TNF-a is a cytokine released from activated astrocytes and microglia, and tightly associated with IL-1β, since the two molecules reciprocally induce their respective release from glia and mutually activate their gene transcription. TNF-a has been shown to increase the mean frequency of AMPA-dependent miniature excitatory postsynaptic currents in hippocampal neurons and to decrease GABAA-mediated inhibitory synaptic strength. These effects are mediated by its ability to activate the recruitment of AMPA receptors lacking the GluR2 subunit at neuronal membranes (thus in a molecular conformation which favors Ca2+ influx

into neurons) and to induce endocytosis of GABAA receptors (Beattie et al. 2002; Stellwagen et al. 2005).

TLR4 is the TLR subtype most extensively studied for its involvement in brain excitability. Cortical rat exposure to lipopolysaccaride (LPS), a prototypical activator of TLR4, induces rapid increases in neuronal excitability leading to seizures (Rodgers et al. 2009), which are prevented by IL-1ra, implicating a role of released IL-1β.

Regarding IL-1β, LTP and long-term depression (LTD) impairment are induced by TLR4 stimu-lation, compatible with cognitive deficits induced in rodents by brain inflammation (Galic et al. 2009; Harre et al. 2008; Spencer et al. 2005). These modifications in brain physiology are dependent on the release of TNF-a and IL-1β from activated glial cells (Riazi et al. 2010). Cognitive deficits are associated with specific changes in NMDA receptor subunit expres-sion in the cortex and hippocampus, predicting modifications in CNS excitability (Galic et al. 2009; Harre et al. 2008; Spencer et al. 2005).

(14)

These fast post-translational effects of inflammatory cytokines are examples of novel path-ways by which inflammatory molecules produced in diseased tissue by glia can modulate neurotransmission and contribute to hyperexcitability and associated neuropathology. In general, the amount and persistence of cytokines in brain tissue appears to be a crucial factor which determines their consequences on neuronal excitability. Another important aspect is the pattern of expression of cytokine receptors in the tissue exposed to inflamma-tion. A typical example is represented by TNF-a, which may have inhibitory or permissive effects on seizures depending both on its brain levels and on the receptor subtypes pre-dominantly activated (Balosso et al. 2005). Moreover, transgenic mice with low to moderate overexpression of TNF-a in astrocytes show decreased susceptibility to seizures (Balosso et al. 2005), whereas mice with high overexpression of TNF-a in astrocytes develop signs of neurologic dysfunction (Akassoglou et al. 1997; Probert et al. 1995).

Functional consequences of astrocyte-mediated brain

inflammation on seizures and epileptogenesis

The role of cytokines released by astrocytes in seizures and epileptogenesis has been inves-tigated at various levels, initially using genetically-modified mice with perturbed cytokine systems and subsequently by pharmacological intervention using receptor antagonists or cytokine synthesis inhibitors, or by injecting the cytokines themselves into rodent brain [re-viewed in (Vezzani et al. 2008a; Vezzani et al. 2011b)].

IL-1β, TNF-a and HMGB1 are among the most studied cytokines for their permissive role in seizures. Mice with upregulation of IL-1ra in astrocytes or lacking IL-1R1 are intrinsically resistant to seizures (Vezzani et al. 2000) and the intracerebral injection of IL-1ra mediates powerful anticonvulsant effects (Vezzani et al. 2000; Vezzani et al. 2002). Because the only action of IL-1ra is to inhibit the effects of IL-1β, these data demonstrate that an endogenous increase in brain IL-1β contributes to seizures. Mice lacking caspase-1, the biosynthetic en-zyme of IL-1β, and therefore unable to release the biologically active form of this cytokine, show decreased seizure susceptibility (Ravizza et al. 2006b). Pharmacological data support a proconvulsant role of IL-1β in several acute and chronic seizure models (Vezzani et al. 1999; Vezzani et al. 2000), as well as in the kindling model of epileptogenesis (Ravizza et al. 2008b). Moreover, recent data showed that inhibition of IL-1β biosynthesis in astrocytes reduces spike-and-wave discharges in rats with genetic absence epilepsy (GAERS) (Akin et al. 2011). Increase in astrocytic IL-1β in the hippocampus due to fever/hyperthermia is in-volved in decreasing the seizure threshold in the immature rodent (Dube et al. 2005; Heida and Pittman 2005).

(15)

or injured neurons (i.e. HMGB1), promotes seizures by activation of neuronal TLR. Seizures, in turn, induce an additional wave of HMGB1 release from activated astrocytes and micro-glia, leading to a vicious positive feedback cycle of seizures and inflammation. This novel pathway may be a crucial mechanism for recurrent seizures [(Maroso et al. 2010); reviewed in (Maroso et al. 2011; Vezzani et al. 2011d); Fig. 2].

Cytokines and other inflammatory mediators have been shown to contribute to both ex-citotoxic and apoptotic neuronal death (Allan et al. 2005), highlighting the possibility that they contribute to seizure-mediated neuronal damage. The deleterious effects of cytokines on neuronal survival involve the production of neurotoxic compounds via autocrine or par-acrine mechanisms (Allan et al. 2005; Vezzani and Baram 2007). Importantly, although cy-tokines can promote neurodegeneration (Fig.2), their effects on the threshold, frequency and duration of seizures are not dependent on cell death (Vezzani et al. 2011c).

Notable examples exist of a dual role of cytokines on neuronal survival in diseased tissue (Allan et al. 2005; Bernardino et al. 2005); it has been shown the ability of cytokines to induce the synthesis of growth factors in astrocytes, to activate antioxidant pathways, man-ganese superoxide dismutase, or calbindin which counteracts the elevation of intracellular Ca2+ induced by cell injuries (Allan et al. 2005), thus promoting cell repair mechanisms. In

this respect, IL-1β and TNF-a can either reduce or exacerbate glutamate receptor–mediated excitotoxicity in organotypic slice cultures, depending on their extracellular concentrations, the length of time the tissue is exposed to these cytokines, and the receptor types activated by these cytokines (Bernardino et al. 2005).

Finally, the possible involvement of inflammatory mediators in epileptogenesis has been suggested by two main lines of evidence: the induction of an inflammatory state in the brain by administering proinflammatory molecules in rodents, or the use of mice that overexpress specific cytokines in astrocytes. This leads to decreased seizure threshold and induces term neurological deficits, particularly if applied to immature rodents, thus suggesting long-term effects of inflammation on brain functions [reviewed by (Ravizza et al. 2011; Riazi et al. 2010)]. In this context, brain inflammation has been implicated in the pathophysiology of several neuropsychiatric conditions (such as depression, memory impairments, and autism spectrum disorder) which are comorbidities of epilepsy, (Vezzani et al. 2011a).

Pharmacological interference with specific inflammatory pathways activated during epi-leptogenesis may reduce the severity and frequency of spontaneous seizures [reviewed in (Ravizza et al. 2011)].

Additionally, cytokines such as IL-1β and HMGB1 released in diseased tissue by parenchy-mal and, in particular, by perivascular astrocytes, can play a major role in BBB breakdown associated with brain inflammation in human and experimental epileptic tissue [reviewed

(16)

by (Friedman et al. 2009)]. The opening of the BBB rapidly activates the innate immune response (Cacheaux, 2002) and the accumulation of albumin in the brain because of BBB damage. Albumin triggers long-lasting hyperexcitability in surrounding tissue by impairing astrocyte capacity to buffer extracellular K+ and glutamate via activation of the TGF-β

path-way [reviewed by (Friedman et al. 2009)].

Finally, it is increasingly recognized that pro-inflammatory molecules released by glia con-tribute to some of the acquired channelopathies described in epilepsy by inducing altera-tions in voltage- and receptor-gated ion channels via either post-translational or transcrip-tional mechanisms [reviewed by (Vezzani et al. 2011b; Viviani et al. 2004)].

Concluding remarks

During the past decade, detailed molecular characterization of astrocytes, in particular re-active astrocytes, demonstrates that these cells are re-active players in the development and progression of the immune/inflammatory response that takes place in epileptic brain tis-sue. Both human and experimental data suggest the activation of specific proinflammatory pathways in astrocytes, which may also recruit neuronal cells and, in some cases, cells of the adaptive immune system. The identification of “harmful” pro-inflammatory pathways contributing to seizure onset and recurrence, as well as to comorbities often associated with epilepsy, highlights the possibility of developing a therapeutic strategy targeting the astrocyte-mediated inflammatory signalings. However, we need to wait for the outcome of clinical studies before we can consider whether this approach is the right strategy. If this is so, it may not only improve control of seizures, but may also act as disease-modifying therapy in patients with epilepsy resistant to conventional antiepileptic drugs.

Acknowledgements

AV is supported by Fondazione Monzino, Fondazione Cariplo and PACE; EA is supported by National Epilepsy Funds (NEF 09-05) and EU FP7 project NeuroGlia (Grant Agreement N° 202167).

(17)

References

1. Abbracchio MP, Ceruti S. 2007. P1 receptors and cytokine secretion. Purinergic Signal 3:13-25.

2. Akassoglou K, Probert L, Kontogeorgos G, Kollias G. 1997. Astrocyte-specific but not neuron-specific transmembrane TNF triggers inflammation and degeneration in the central nervous system of transgenic mice. J Immunol 158:438-45.

3. Akin D, Ravizza T, Maroso M, Carcak N, Eryigit T, Vanzulli I, Aker RG, Vezzani A, Yilmaz FO. 2011. IL-1βeta is induced in reactive astrocytes in the somatosensory cortex of rats with genetic absence epilepsy at the onset of spike-and-wave discharges, and contributes to their occurrence. Neurobiol Dis. 44(3):259-69.

4. Allan SM, Tyrrell PJ, Rothwell NJ. 2005. Interleukin-1 and neuronal injury. Nat Rev Im-munol 5:629-40.

5. Aloisi F, Ria F, Penna G, Adorini L. 1998. Microglia are more efficient than astrocytes in antigen processing and in Th1 but not Th2 cell activation. J Immunol 160:4671-80.

6. Aloisi F, Ria F, Adorini L. 2000a. Regulation of T-cell responses by CNS antigen-presenting cells: different roles for microglia and astrocytes. Immunol Today 21:141-7.

7. Aloisi F, Serafini B, Adorini L. 2000b. Glia-T cell dialogue. J Neuroimmunol 107:111-7.

8. Aloisi F. 2001. Immune function of microglia. Glia 36:165-79.

9. Amor S, Puentes F, Baker D, van der Valk P. 2010. Inflammation in neurodegenerative diseases. Immunology 129:154-69.

10. Andjelkovic AV, Kerkovich D, Pachter JS. 2000. Monocyte:astrocyte interactions regulate MCP-1 expression in both cell types. J Leukoc Biol 68:545-52.

11. Aronica E, Boer K, Becker A, Redeker S, Spliet WG, van Rijen PC, Wittink F, Breit T, Wad-man WJ, Lopes da Silva FH and others. 2008. Gene expression profile analysis of epilep-sy-associated gangliogliomas. Neuroscience 151:272-92.

12. Aronica E, Boer K, van Vliet EA, Baayen JC, Redeker S, Spliet WGM, Lopes da Silva FH, Wadman WJ, Troost D, Gorter JA. 2007. Complement activation in experimental and human temporal lobe epilepsy. Neurobiol Dis 26:497-511.

13. Aronica E, Crino PB. 2011. Inflammation in epilepsy: clinical observations. Epilepsia 52 Suppl 3:26-32.

14. Aronica E, Fluiter K, Iyer A, Zurolo E, Vreijling J, van Vliet EA, Baayen JC, Gorter JA. 2010. Expression pattern of miR-146a, an inflammation-associated microRNA, in experimen-tal and human temporal lobe epilepsy. Eur J Neurosci 31:1100-7.

15. Aronica E, Gorter J. 2007. Gene Expression Profile in Temporal Lobe Epilepsy. Neurosci-entist 13:1-9.

(18)

16. Aronica E, Gorter JA, Ijlst-Keizers H, Rozemuller AJ, Yankaya B, Leenstra S, Troost D. 2003. Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J Neurosci 17:2106-18.

17. Aronica E, Gorter JA, Rozemuller AJ, Yankaya B, Troost D. 2005a. Activation of metabo-tropic glutamate receptor 3 enhances interleukin (IL)-1beta-stimulated release of IL-6 in cultured human astrocytes. Neuroscience 130:927-33.

18. Aronica E, Gorter JA, Rozemuller AJ, Yankaya B, Troost D. 2005b. Interleukin-1 beta down-regulates the expression of metabotropic glutamate receptor 5 in cultured hu-man astrocytes. J Neuroimmunol 160:188-94.

19. Aronica E, van Vliet EA, Mayboroda OA, Troost D, da Silva FH, Gorter JA. 2000. Upregu-lation of metabotropic glutamate receptor subtype mGluR3 and mGluR5 in reactive astrocytes in a rat model of mesial temporal lobe epilepsy. Eur J Neurosci 12:2333-44.

20. Aronica E, Zurolo E, Iyer A, de Groot M, Anink J, Carbonell C, van Vliet EA, Baayen JC, Boison D, Gorter JA. 2011b. Upregulation of adenosine kinase in astrocytes in experi-mental and human temporal lobe epilepsy. Epilepsia.

21. Balosso S, Ravizza T, Perego C, Peschon J, Campbell IL, De Simoni MG, Vezzani A. 2005. Tumor necrosis factor-alpha inhibits seizures in mice via p75 receptors. Ann Neurol 57:804-12.

22. Barnum SR, Jones JL. 1995. Differential regulation of C3 gene expression in human as-troglioma cells by interferon-gamma and interleukin-1 beta. Neurosci Lett 197:121-4.

23. Bauer B, Hartz AM, Pekcec A, Toellner K, Miller DS, Potschka H. 2008. Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cy-clooxygenase-2 signaling. Mol Pharmacol 73:1444-53.

24. Bauer J, Elger CE, Hans VH, Schramm J, Urbach H, Lassmann H, Bien CG. 2007. Astrocytes are a specific immunological target in Rasmussen’s encephalitis. Ann Neurol 62:67-80.

25. Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. 2002. Cell death in models of spinal cord injury. Prog Brain Res 137:37-47.

26. Becher B, Prat A, Antel JP. 2000. Brain-immune connection: immuno-regulatory proper-ties of CNS-resident cells. Glia 29:293-304.

27. Bellinger FP, Madamba S, Siggins GR. 1993. Interleukin 1 beta inhibits synaptic strength and long-term potentiation in the rat CA1 hippocampus. Brain Res 628:227-34.

28. Benarroch EE. 2007. Tissue plasminogen activator: beyond thrombolysis. Neurology 69:799-802.

29. Bernardino L, Ferreira R, Cristovao AJ, Sales F, Malva JO. 2005. Inflammation and neuro-genesis in temporal lobe epilepsy. Curr Drug Targets CNS Neurol Disord 4:349-60.

(19)

Kollias G, Meldolesi J and others. 2001. CXCR4-activated astrocyte glutamate release via TNFalpha: amplification by microglia triggers neurotoxicity. Nat neurosci 4:702-10.

31. Bianchi ME, Manfredi AA. 2009. Immunology. Dangers in and out. Science 323:1683-4.

32. Blumcke I, Thom M, Aronica E, Armstrong DD, Vinters HV, Palmini A, Jacques TS, Avan-zini G, Barkovich AJ, Battaglia G and others. 2011. The clinico-pathological spectrum of Focal Cortical Dysplasias : a consensus classification proposed by an ad hoc Task Force of the ILAE Diagnostics Commission. Epilepsia 52: 158–174.

33. Blümcke I, Vinters HV, Armstrong D, Aronica E, Thom M, Spreafico R. 2009. Malforma-tions of Cortical Development and Epilepsies: Neuropathological findings with empha-sis on Focal Cortical Dysplasia. Epileptic Disord 11:181-93.

34. Boer K, Crino PB, Gorter JA, Nellist M, Jansen FE, Spliet WG, van Rijen PC, Wittink F, Breit T, Troost D and others. 2010. Gene Expression Analysis of Tuberous Sclerosis Complex Cortical Tubers Reveals Increased Expression of Adhesion and Inflammatory Factors. Brain Pathol 20:704-719.

35. Boer K, Troost D, Spliet WG, van Rijen PC, Gorter JA, Aronica E. 2008. Cellular distribu-tion of vascular endothelial growth factor A (VEGFA) and B (VEGFB) and VEGF receptors 1 and 2 in focal cortical dysplasia type IIB. Acta Neuropat 115:683-96.

36. Boison D. 2010. Adenosine dysfunction and adenosine kinase in epileptogenesis. Open Neurosci J 4:93-101.

37. Bonifati DM, Kishore U. 2007. Role of complement in neurodegeneration and neuroin-flammation. Mol Immunol 44:999-1010.

38. Boon CJ, van de Kar NC, Klevering BJ, Keunen JE, Cremers FP, Klaver CC, Hoyng CB, Daha MR, den Hollander AI. 2009. The spectrum of phenotypes caused by variants in the CFH gene. Mol Immunol 46:1573-94.

39. Byrnes KR, Loane DJ, Faden AI. 2009. Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurotherapeutics 6:94-107.

40. Carmignoto G, Fellin T. 2006. Glutamate release from astrocytes as a non-synaptic mechanism for neuronal synchronization in the hippocampus. J Physiol 99:98-102.

41. Casamenti F, Prosperi C, Scali C, Giovannelli L, Colivicchi MA, Faussone-Pellegrini MS, Pepeu G. 1999. Interleukin-1beta activates forebrain glial cells and increases nitric oxide production and cortical glutamate and GABA release in vivo: implications for Alzhei-mer’s disease. Neuroscience 91:831-42.

42. Chew LJ, Takanohashi A, Bell M. 2006. Microglia and inflammation: impact on develop-mental brain injuries. Ment. Retard. Dev. Disabil. Res. Rev. 12:105-12.

43. Cohly HH, Panja A. 2005. Immunological findings in autism. Int Rev Neurobiol 71:317-41.

(20)

44. Cornet A, Bettelli E, Oukka M, Cambouris C, Avellana-Adalid V, Kosmatopoulos K, Liblau RS. 2000. Role of astrocytes in antigen presentation and naive T-cell activation. J Neu-roimmunol 106:69-77.

45. Crack PJ, Bray PJ. 2007. Toll-like receptors in the brain and their potential roles in neuro-pathology. Immunol Cell Biol 85:476-80.

46. Crespel A, Coubes P, Rousset MC, Brana C, Rougier A, Rondouin G, Bockaert J, Baldy-Moulinier M, Lerner-Natoli M. 2002. Inflammatory reactions in human medial temporal lobe epilepsy with hippocampal sclerosis. Brain Res 952:159-69.

47. Cross AH, Ku G. 2000. Astrocytes and central nervous system endothelial cells do not express B7-1 (CD80) or B7-2 (CD86) immunoreactivity during experimental autoim-mune encephalomyelitis. J Neuroimmunol 110:76-82.

48. Cui JG, Li YY, Zhao Y, Bhattacharjee S, Lukiw WJ. 2010. Differential regulation of interleu-kin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kap-paB in stressed human astroglial cells and in Alzheimer disease. J Biol Chem 285:38951-60.

49. D’Antoni S, Berretta A, Bonaccorso CM, Bruno V, Aronica E, Nicoletti F, Catania MV. 2008. Metabotropic glutamate receptors in glial cells. Neurochem Res 33:2436-43.

50. De Keyser J, Zeinstra E, Frohman E. 2003. Are astrocytes central players in the patho-physiology of multiple sclerosis? Arch Neurol 60:132-6.

51. De Simoni MG, Perego C, Ravizza T, Moneta D, Conti M, Marchesi F, De Luigi A, Garattini S, Vezzani A. 2000. Inflammatory cytokines and related genes are induced in the rat hip-pocampus by limbic status epilepticus. Eur J Neurosci 12:2623-33.

52. Desjardins P, Sauvageau A, Bouthillier A, Navarro D, Hazell AS, Rose C, Butterworth RF. 2003. Induction of astrocytic cyclooxygenase-2 in epileptic patients with hippocampal sclerosis. Neurochem Int 42:299-303.

53. Dinarello CA. 2004. Infection, fever, and exogenous and endogenous pyrogens: some concepts have changed. J Endotoxin Res 10:201-22.

54. Dong Y, Benveniste EN. 2001. Immune function of astrocytes. Glia 36:180-90.

55. Dube C, Vezzani A, Behrens M, Bartfai T, Baram TZ. 2005. Interleukin-1beta contributes to the generation of experimental febrile seizures.[see comment. Ann Neurol 57:152-5.

56. Farina C, Aloisi F, Meinl E. 2007. Astrocytes are active players in cerebral innate immu-nity. Trends Immunol 28:138-45.

57. Farrell MA, Droogan O, Secor DL, Poukens V, Quinn B, Vinters HV. 1995. Chronic en-cephalitis associated with epilepsy: immunohistochemical and ultrastructural studies. Acta Neuropathol 89:313-21.

(21)

Plaw-inski L, MacKenzie ET, Bu G, Buisson A, Vivien D. 2004. Is tissue-type plasminogen acti-vator a neuromodulator? Molecul Cel Neurosci 25:594-601.

59. Fischer W, Appelt K, Grohmann M, Franke H, Norenberg W, Illes P. 2009. Increase of intracellular Ca2+ by P2X and P2Y receptor-subtypes in cultured cortical astroglia of the rat. Neuroscience 160:767-83.

60. Friedman A, Dingledine R. 2011. Molecular cascades that mediate the influence of in-flammation on epilepsy. Epilepsia 52 Suppl 3:33-9.

61. Friedman A, Kaufer D, Heinemann U. 2009. Blood-brain barrier breakdown-inducing astrocytic transformation: novel targets for the prevention of epilepsy. Epilepsy Res 85:142-9.

62. Galic MA, Riazi K, Henderson AK, Tsutsui S, Pittman QJ. 2009. Viral-like brain inflamma-tion during development causes increased seizure susceptibility in adult rats. Neurobiol Dis 36:343-51.

63. Gantier MP. 2010. New perspectives in MicroRNA regulation of innate immunity. J Inter-feron Cytokine Res 30:283-9.

64. Girvin AM, Gordon KB, Welsh CJ, Clipstone NA, Miller SD. 2002. Differential abilities of central nervous system resident endothelial cells and astrocytes to serve as inducible antigen-presenting cells. Blood 99:3692-701.

65. Glynn MW, Elmer BM, Garay PA, Liu XB, Needleman LA, El-Sabeawy F, McAllister AK. 2011. MHCI negatively regulates synapse density during the establishment of cortical connections. Nat Neurosci 14:442-51.

66. Gobin SJ, Montagne L, Van Zutphen M, Van Der Valk P, Van Den Elsen PJ, De Groot CJ. 2001. Upregulation of transcription factors controlling MHC expression in multiple scle-rosis lesions. Glia 36:68-77.

67. Gomes CV, Kaster MP, Tome AR, Agostinho PM, Cunha RA. 2011. Adenosine receptors and brain diseases: neuroprotection and neurodegeneration. Biochim Biophys Acta 1808:1380-99.

68. Gorter JA, Van Vliet E, Aronica E, Rauwerda H, Breit T, Lopes da Silva FH, Wadman WJ. 2006. Potential New Antiepileptogenic Targets Indicated by Microarray Analysis in a Rat Model for Temporal Lobe Epilepsy. J Neurosci 26:11083-110.

69. Graeber MB, Streit WJ. 1990. Microglia: immune network in the CNS. Brain Pathol 1:2-5.

70. Gravanis I, Tsirka SE. 2005. Tissue plasminogen activator and glial function. GLIA 49:177-83.

71. Griffiths MR, Neal JW, Fontaine M, Das T, Gasque P. 2009. Complement factor H, a marker of self protects against experimental autoimmune encephalomyelitis. J Immu-nol 182:4368-77.

(22)

72. Hajishengallis G, Lambris JD. 2010. Crosstalk pathways between Toll-like receptors and the complement system. Trends Immunol 31:154-163.

73. Harre EM, Galic MA, Mouihate A, Noorbakhsh F, Pittman QJ. 2008. Neonatal inflamma-tion produces selective behavioural deficits and alters N-methyl-D-aspartate receptor subunit mRNA in the adult rat brain. Eur J Neurosci 27:644-53.

74. Hasko G, Pacher P, Vizi ES, Illes P. 2005. Adenosine receptor signaling in the brain im-mune system. Trends Pharmacol Sci 26:511-6.

75. Hayakawa K, Arai K, Lo EH. 2010. Role of ERK map kinase and CRM1 in IL-1βeta-stimulated release of HMGB1 from cortical astrocytes. Glia 58:1007-15.

76. Heida JG, Pittman QJ. 2005. Causal links between brain cytokines and experimental febrile convulsions in the rat. Epilepsia 46.:1906-1913.

77. Hewett SJ, Csernansky CA, Choi DW. 1994. Selective potentiation of NMDA-induced neuronal injury following induction of astrocytic iNOS. Neuron 13:487-94.

78. Holtman L, van Vliet EA, van Schaik R, Queiroz CM, Aronica E, Gorter JA. 2009. Effects of SC58236, a selective COX-2 inhibitor, on epileptogenesis and spontaneous seizures in a rat model for temporal lobe epilepsy. Epilepsy Res 84:56-66.

79. Hu S, Sheng WS, Ehrlich LC, Peterson PK, Chao CC. 2000. Cytokine effects on glutamate uptake by human astrocytes. Neuroimmunomodulation 7:153-9.

80. Ivens S, Kaufer D, Flores LP, Bechmann I, Zumsteg D, Tomkins O, Seiffert E, Heinemann U, Friedman A. 2007. TGF-beta receptor-mediated albumin uptake into astrocytes is involved in neocortical epileptogenesis. Brain 130:535-47.

81. Iyer AM, Zurolo E, Boer K, Baayen JC, Giangaspero F, Arcella A, Di Gennaro GC, Esposito V, Spliet WG, van Rijen PC and others. 2010. Tissue plasminogen activator and urokinase plasminogen activator in human epileptogenic pathologies. Neuroscience 167:929-45.

82. Kawai T, Akira S. 2007. Signaling to NF-κB by Toll-like receptors Trends in Molecular Medicine 13:460-469.

83. Khurgel M, Switzer RC, 3rd, Teskey GC, Spiller AE, Racine RJ, Ivy GO. 1995. Activation of astrocytes during epileptogenesis in the absence of neuronal degeneration. Neurobiol Dis 2:23-35.

84. Kielian T. 2006. Toll-like receptors in central nervous system glial inflammation and ho-meostasis. J Neurosci Res 83:711-30.

85. Kim SK, Wang KC, Hong SJ, Chung CK, Lim SY, Kim YY, Chi JG, Kim CJ, Chung YN, Kim HJ and others. 2003. Gene expression profile analyses of cortical dysplasia by cDNA arrays. Epilepsy Res 56:175-83.

86. Kulkarni SK, Dhir A. 2009. Cyclooxygenase in epilepsy: from perception to application. Drugs of today 45:135-54.

(23)

87. Lai AY, Swayze RD, El-Husseini A, Song C. 2006. Interleukin-1 beta modulates AMPA receptor expression and phosphorylation in hippocampal neurons. JJ Neuroimmunol 175:97-106.

88. Liu JS, John GR, Sikora A, Lee SC, Brosnan CF. 2000. Modulation of interleukin-1beta and tumor necrosis factor alpha signaling by P2 purinergic receptors in human fetal astro-cytes. J Neurosci 20:5292-9.

89. Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, Iyer AM, Rossetti C, Molteni M, Man-fredi AA, E. BM and others. 2010. Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nature Med 1 16:413-419 .

90. Maroso M, Balosso S, Ravizza T, Liu J, Bianchi ME, Vezzani A. 2011. Interleukin-1 type 1 receptor/Toll-like receptor signalling in epilepsy: the importance of IL-1βeta and high-mobility group box 1. J Int Med 270:319-26.

91. Matute C, Cavaliere F. 2011. Neuroglial interactions mediated by purinergic signalling in the pathophysiology of CNS disorders. Semin Cell Dev Biol 22:252-9.

92. McNaull BB, Todd S, McGuinness B, Passmore AP. 2010. Inflammation and anti-inflam-matory strategies for Alzheimer’s disease--a mini-review. Gerontology 56:3-14.

93. Meeuwsen S, Persoon-Deen C, Bsibsi M, Ravid R, van Noort JM. 2003. Cytokine, chemokine and growth factor gene profiling of cultured human astrocytes after expo-sure to proinflammatory stimuli. Glia 43:243-53.

94. Morin-Brureau M, Lebrun A, Rousset MC, Fagni L, Bockaert J, de Bock F, Lerner-Natoli M. 2011. Epileptiform activity induces vascular remodeling and zonula occludens 1 downregulation in organotypic hippocampal cultures: role of VEGF signaling pathways. J Neurosci 31:10677-88.

95. Muller S, Ronfani L, Bianchi ME. 2004. Regulated expression and subcellular localization of HMGB1, a chromatin protein with a cytokine function. JJ Int Med 255:332-43.

96. O’Donnell E, Vereker E, Lynch MA. 2000. Age-related impairment in LTP is accompanied by enhanced activity of stress-activated protein kinases: analysis of underlying mecha-nisms. Eur J Neurosci 12:345-52.

97. O’Neill LA. 2008. The interleukin-1 receptor/Toll-like receptor superfamily: 10 years of progress. Immunol Rev 226:10-8.

98. O’Neill LA, Dinarello CA. 2000. The IL-1 receptor/toll-like receptor superfamily: crucial receptors for inflammation and host defense. Immunol Today 21:206-9.

99. Oeckinghaus A, Hayden MS, Ghosh S. 2011. Crosstalk in NF-kappaB signaling pathways. Nat immunol 12:695-708.

(24)

101. Panenka W, Jijon H, Herx LM, Armstrong JN, Feighan D, Wei T, Yong VW, Ransohoff RM, MacVicar BA. 2001. P2X7-like receptor activation in astrocytes increases chemokine monocyte chemoattractant protein-1 expression via mitogen-activated protein kinase. J Neurosci 21:7135-42.

102. Parker WE, Orlova KA, Heuer GG, Baybis M, Aronica E, Frost M, Wong M, Crino PB. 2011. Enhanced epidermal growth factor, hepatocyte growth factor, and vascular endothelial growth factor expression in tuberous sclerosis complex. Am J Pathol 178:296-305.

103. Pedrazzi M, Patrone M, Passalacqua M, Ranzato E, Colamassaro D, Sparatore B, Pon-tremoli S, Melloni E. 2007. Selective proinflammatory activation of astrocytes by high-mobility group box 1 protein signaling. J Immunol 179:8525-32.

104. Plata-Salaman CR, ffrench-Mullen JM. 1994. Interleukin-1 beta inhibits Ca2+ channel currents in hippocampal neurons through protein kinase C. Eur J Pharmacol 266:1-10.

105. Probert L, Plows D, Kontogeorgos G, Kollias G. 1995. The type I interleukin-1 recep-tor acts in series with tumor necrosis facrecep-tor (TNF) to induce arthritis in TNF-transgenic mice. Eur J Immunol 25:1794-7.

106. Quinn SR, O’Neill LA. 2011. A trio of microRNAs that control Toll-like receptor signalling. Int Immunol 23:421-5.

107. Ravizza T, Balosso S, Vezzani A. 2011. Inflammation and prevention of epileptogenesis. Neurosci lett 497:223-30.

108. Ravizza T, Boer K, Redeker S, Spliet WG, van Rijen PC, Troost D, Vezzani A, Aronica E. 2006a. The IL-1βeta system in epilepsy-associated malformations of cortical develop-ment. Neurobiol Dis 24:128-43.

109. Ravizza T, Gagliardi B, Noe F, Boer K, Aronica E, Vezzani A. 2008a. Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimen-tal models and human temporal lobe epilepsy. Neurobiol Dis 29:142-60.

110. Ravizza T, Lucas SM, Balosso S, Bernardino L, Ku G, Noe’ F, Malva J, Rande JC, Allan S, Vezzani A. 2006b. Inactivation of caspase-1 in rodent brain: a novel anticonvulsive strat-egy. Epilepsia 47:1160-8.

111. Ravizza T, Noe F, Zardoni D, Vaghi V, Sifringer M, Vezzani A. 2008b. Interleukin Convert-ing Enzyme inhibition impairs kindlConvert-ing epileptogenesis in rats by blockConvert-ing astrocytic IL-1βeta production. Neurobiol Dis 31:327-33.

112. Riazi K, Galic MA, Pittman QJ. 2010. Contributions of peripheral inflammation to seizure susceptibility: cytokines and brain excitability. Epilepsy Research 89:34-42.

113. Rigau V, Morin M, Rousset MC, de Bock F, Lebrun A, Coubes P, Picot MC, Baldy-Moulin-ier M, Bockaert J, Crespel A and others. 2007a. Angiogenesis is associated with blood-brain barrier permeability in temporal lobe epilepsy. Brain 130:1942-56.

(25)

114. Rodgers KM, Hutchinson MR, Northcutt A, Maier SF, Watkins LR, Barth DS. 2009. The cortical innate immune response increases local neuronal excitability leading to sei-zures. Brain 132:2478-86.

115. Samadani U, Judkins AR, Akpalu A, Aronica E, Crino PB. 2007. Differential cellular gene expression in ganglioglioma. Epilepsia 48:646-53.

116. Schneider H, Pitossi F, Balschun D, Wagner A, del Rey A, Besedovsky HO. 1998. A neu-romodulatory role of interleukin-1beta in the hippocampus. Ann N Y Acad Sci, 95:7778-83.

117. Schoch HJ, Fischer S, Marti HH. 2002. Hypoxia-induced vascular endothelial growth fac-tor expression causes vascular leakage in the brain. Brain 125:2549-57.

118. Schwartz M, Kipnis J. 2011. A conceptual revolution in the relationships between the brain and immunity. Brain Behav, Immun 25:817-9.

119. Seifert G, Carmignoto G, Steinhauser C. 2010. Astrocyte dysfunction in epilepsy. Brain Res Rev 63:212-21.

120. Sheedy FJ, O’Neill LA. 2008. Adding fuel to fire: microRNAs as a new class of mediators of inflammation. Ann Rheum Dis 67 Suppl 3:iii50-5.

121. Sheng WS, Hu S, Min X, Cabral GA, Lokensgard JR, Peterson PK. 2005. Synthetic can-nabinoid WIN55,212-2 inhibits generation of inflammatory mediators by IL-1βeta-stimulated human astrocytes. GLIA 49:211-9.

122. Sofroniew MV, Vinters HV. 2010. Astrocytes: biology and pathology. Acta Neuropatho-logica 119:7-35.

123. Sonkoly E, Stahle M, Pivarcsi A. 2008. MicroRNAs and immunity: novel players in the regulation of normal immune function and inflammation. Sem Cancer Biol 18:131-40.

124. Sosunov AA, Wu X, Weiner HL, Mikell CB, Goodman RR, Crino PD, McKhann GM, 2nd. 2008. Tuberous sclerosis: a primary pathology of astrocytes? Epilepsia 49 Suppl 2:53-62.

125. Spencer SJ, Heida JG, Pittman QJ. 2005. Early life immune challenge--effects on behav-ioural indices of adult rat fear and anxiety. Behav Brain Res 164:231-8.

126. Stellwagen D, Beattie EC, Seo JY, Malenka RC. 2005. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J Neurosci 25:3219-28.

127. Steward O, Torre ER, Tomasulo R, Lothman E. 1992. Seizures and the regulation of astro-glial gene expression. Epilepsy Res Suppl 7:197-209.

128. Taganov KD, Boldin MP, Chang KJ, Baltimore D. 2006. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune re-sponses. PNAS 103:12481-6.

(26)

129. Tian GF, Azmi H, Takano T, Xu Q, Peng W, Lin J, Oberheim N, Lou N, Wang X, Zielke HR and others. 2005. An astrocytic basis of epilepsy. Nat Med 11:973-81.

130. Traugott U, Raine CS. 1985. Multiple sclerosis. Evidence for antigen presentation in situ by endothelial cells and astrocytes. J Neurol Sci 69:365-70.

131. van Vliet EA, da Costa Araújo S, Redeker S, van Schaik R, Aronica E, Gorter JA. 2007. Long-lasting increased permeability of the blood-brain barrier may contribute to sei-zure progression in temporal lobe epilepsy. Brain 130:521-534.

132. van Vliet EA, Zibell G, Pekcec A, Schlichtiger J, Edelbroek PM, Holtman L, Aronica E, Gorter JA, Potschka H. 2009. COX-2 inhibition controls P-glycoprotein expression and promotes brain delivery of phenytoin in chronic epileptic rats. Neuropharmacol 58:404-12.

133. Veerhuis R, Janssen I, De Groot CJ, Van Muiswinkel FL, Hack CE, Eikelenboom P. 1999. Cytokines associated with amyloid plaques in Alzheimer’s disease brain stimulate hu-man glial and neuronal cell cultures to secrete early complement proteins, but not C1-inhibitor. Exp Neurol 160:289-99.

134. Vezzani A, Aronica E, Mazarati A, Pittman QJ. 2011a. Epilepsy and brain inflammation. Exp Neurol [Epub ahead of print]

135. Vezzani A, Balosso S, Maroso M, Zardoni D, Noé F, Ravizza T. 2010. ICE/caspase 1 inhibi-tors and IL-1βeta receptor antagonists as potential therapeutics in epilepsy. Curr Opin Investig Drugs 11:43-50.

136. Vezzani A, Balosso S, Ravizza T. 2008a. The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun 22:797-803.

137. Vezzani A, Baram TZ. 2007. New roles for interleukin-1 Beta in the mechanisms of epi-lepsy. Epilepsy Curr / AES 7:45-50.

138. Vezzani A, Conti M, De Luigi A, Ravizza T, Moneta D, Marchesi F, De Simoni MG. 1999. Interleukin-1beta immunoreactivity and microglia are enhanced in the rat hippocam-pus by focal kainate application: functional evidence for enhancement of electrographic seizures. J Neurosci 19:5054-65.

139. Vezzani A, French J, Bartfai T, Baram TZ. 2011b. The role of inflammation in epilepsy. Nature Rev Neurol 7:31-40.

140. Vezzani A, Maroso M, Balosso S, Sanchez MA, Bartfai T. 2011c. IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hy-perexcitability and seizures. Brain Behav Immun 25:1281-9.

141. Vezzani A, Maroso M, Balosso S, Sanchez MA, Bartfai T. 2011d. IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hy-perexcitability and seizures. Brain Behav Immun.

(27)

142. Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J and others. 2000. Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. PNAS 97:11534-9.

143. Vezzani A, Ravizza T, Balosso S, Aronica E. 2008b. Glia as a source of cytokines: implica-tions for neuronal excitability and survival. Epilepsia 49 Suppl 2:24-32.

144. Viviani B, Bartesaghi S, Corsini E, Galli CL, Marinovich M. 2004. Cytokines role in neuro-degenerative events. Toxicol Lett 149:85-9.

145. Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T, Binaglia M, Corsini E, Di Luca M, Galli CL and others. 2003. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J. Neurosci 23:8692-700.

146. Weber F, Meinl E, Aloisi F, Nevinny-Stickel C, Albert E, Wekerle H, Hohlfeld R. 1994. Hu-man astrocytes are only partially competent antigen presenting cells. Possible implica-tions for lesion development in multiple sclerosis. Brain 117 ( Pt 1):59-69.

147. Wetherington J, Serrano G, Dingledine R. 2008. Astrocytes in the epileptic brain. Neu-ron 58:168-78.

148. Wieser HG. 2004. ILAE Commission Report. Mesial temporal lobe epilepsy with hip-pocampal sclerosis. Epilepsia 45:695-714.

149. Wong M. 2008. Mechanisms of epileptogenesis in tuberous sclerosis complex and re-lated malformations of cortical development with abnormal glioneuronal proliferation. Epilepsia 49:8-21.

150. Wu Y, Wang X, Mo X, Xi Z, Xiao F, Li J, Zhu X, Luan G, Wang Y, Li Y and others. 2008. Ex-pression of monocyte chemoattractant protein-1 in brain tissue of patients with intrac-table epilepsy. Clin Neuropathol 27:55-63.

151. Ye ZC, Sontheimer H. 1996. Cytokine modulation of glial glutamate uptake: a possible involvement of nitric oxide. Neuroreport 7:2181-5.

152. Zurolo E, Iyer A, Maroso M, Carbonell C, Anink JJ, Ravizza T, Fluiter K, Spliet WG, van Rijen PC, Vezzani A and others. 2011. Activation of toll-like receptor, RAGE and HMGB1 signalling in malformations of cortical development. Brain 134:1015-32.

Referenties

GERELATEERDE DOCUMENTEN

An important question for this work was how can the TPCK framework be applied to help in integrating educational EMRs into undergraduate health informatics curricula in order to

Although GitHub expects users to use Gists to in- stantly share code snippets, people are actually using Gists for various purposes: some people use it to create components that can

The transport activity of synthetic ion channel candidates are tested by one (or more) of three general methods: vesicle assays, bilayer clamp experiments, or biological

were synthesized to compare head group effects on cation tran sp o rt.. properties, while different macrocycles were used in th e backbone

The Town of Qualicum Beach (the “Town”), a small, primarily residential waterfront community, located on eastern Vancouver Island, copes with shoreline erosion and sea level rise,

As in the exogenously expressing MEFs, NHA-MAF1b from parasites strongly coimmunoprecipitated MIC60, SAM50, and MIC19 but did not coimmunoprecipitate the abundant cytosolic

Figure 5(a) shows the averaged Raman spectra of the sample prepared with 400 ng/mL AcAm, the blank sample without AcAm, and their difference spectrum (400 ng/mL AcAm –

Trace elements in magnetite from porphyry Cu–Mo–Au deposits in British Columbia, Canada... Trace element concentrations 29