1563

J Physiol 594.6 (2016) pp 1563–1577

Neuroscience

SYMPOSIUM REVIEW

Mechanisms of microglial activation in models of inflammation and hypoxia: Implications for chronic intermittent hypoxia Elizabeth A. Kiernan1 , Stephanie M. C. Smith1 , Gordon S. Mitchell2 and Jyoti J. Watters1 1 2

Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA

The Journal of Physiology

Peripheral inflammation

A

B

1

Direct diffusion BBB breakdown Vagal transmission

Direct diffusion BBB breakdown Vagal transmission

Reactive oxygen species Intermittent hypoxia Reactive oxygen species

3

2

C Inflammation Microglial activation

Neuron death

D DAMPs

Abstract Chronic intermittent hypoxia (CIH) is a hallmark of sleep apnoea, a condition associated with diverse clinical disorders. CIH and sleep apnoea are characterized by increased reactive oxygen species formation, peripheral and CNS inflammation, neuronal death and neurocognitive deficits. Few studies have examined the role of microglia, the resident CNS immune cells, in models of CIH. Thus, little is known concerning their direct contributions to neuropathology or the cellular mechanisms regulating their activities during or following pathological CIH. In this review, we identify gaps in knowledge regarding CIH-induced microglial activation, and propose mechanisms based on data from related models of hypoxia and/or hypoxia–reoxygenation. CIH

Gordon Mitchell is a Professor of Neuroscience in the Department of Physical Therapy and Director of the Center for Respiratory Research and Rehabilitation at the University of Florida in Gainesville. His research interests focus on fundamental mechanisms of spinal respiratory plasticity induced by acute exposures to intermittent hypoxia, and translation of those findings to treat devastating clinical disorders that compromise breathing and non-respiratory movements. Jyoti Watters is a Professor of Pharmacology in the Department of Comparative Biosciences at the University of Wisconsin-Madison. She studies cellular signalling and epigenetic mechanisms that regulate transitions between microglial inflammatory and beneficial activities to harness and manipulate them for therapeutic benefit. She uses rodent models of neuroinflammation induced by exposure to chronic intermittent hypoxia.

E. A. Kiernan and S. M. C. Smith contributed equally to this work. This was presented at the 1st PanAmerican Congress of Physiological Sciences ‘Physiology without Boarders’, Iguassu Falls, Brazil, August 2–4 2014.  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

DOI: 10.1113/JP271502

1564

E. A. Kiernan and others

J Physiol 594.6

may directly affect microglia, or may have indirect effects via the periphery or other CNS cells. Peripheral inflammation may indirectly activate microglia via entry of pro-inflammatory molecules into the CNS, and/or activation of vagal afferents that trigger CNS inflammation. CIH-induced release of damage-associated molecular patterns from injured CNS cells may also activate microglia via interactions with pattern recognition receptors expressed on microglia. For example, Toll-like receptors activate mitogen-activated protein kinase/transcription factor pathways required for microglial inflammatory gene expression. Although epigenetic effects from CIH have not yet been studied in microglia, potential epigenetic mechanisms in microglial regulation are discussed, including microRNAs, histone modifications and DNA methylation. Epigenetic effects can occur during CIH, or long after it has ended. A better understanding of CIH effects on microglial activities may be important to reverse CIH-induced neuropathology in patients with sleep disordered breathing. (Received 30 August 2015; accepted after revision 16 January 2016; first published online 18 February 2016) Correspondiing author J. J. Watters: 2015 Linden Drive, Madison, WI 53706, USA. Email: [email protected] Abstract figure legend Putative mechanisms whereby CIH may contribute to CNS inflammation. Schematic diagram depicting three independent, but interacting, mechanisms of CIH-induced inflammation and neuropathology, and potential interactions between them. CIH increases reactive oxygen species with subsequent peripheral inflammation (1), neuronal injury (2) and microglial activation (3). Peripheral inflammation can cause CNS inflammation via diffusion of inflammatory molecules across an intact or compromised blood–brain barrier (BBB), and/or via vagal afferent neuron activation leading to secondary inflammation in the CNS, and pro-inflammatory microglial activities (A). Peripheral inflammation can induce neuronal injury or cell death by similar mechanisms (B), and/or by microglial production of pro-inflammatory or neurotoxic molecules (C). Neuron–microglial communication propagates neuroinflammation with damaged neurons or glia releasing damage-associated molecular patterns (DAMPs) into the extracellular space where they elicit microglial inflammatory activities via activation of pattern recognition receptors (e.g. Toll-like receptors) or scavenger receptors (D). Abbreviations CIH, chronic intermittent hypoxia; CSA, central sleep apnoea; DAMP, damage-associated molecular pattern; HRO, hypoxia–reoxygenation; LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; OSA, obstructive sleep apnoea; PRR, pattern recognition receptor; TLR, Toll-like receptor.

Introduction

Chronic intermittent hypoxia (CIH) is a hallmark of sleep-disordered breathing. Although sleep apnoea (and CIH) is highly prevalent in otherwise normal individuals (Peppard et al. 2013), it is even more prevalent in clinical disorders of the central nervous system, including traumatic brain and spinal cord injury (Tran et al. 2010), ischaemic injury (stroke; Park et al. 2011), and genetic (e.g. Down’s syndrome; Marcus et al. 1991) or neurodegenerative diseases (e.g. Alzheimer’s and Parkinson’s diseases, multiple sclerosis, amyotrophic lateral sclerosis; Hoch et al. 1986; Manon-Espaillat et al. 1989; Atalaia et al. 2007; Bombois et al. 2010). Obstructive (OSA) and central (CSA) sleep apnoea cause morbidities such as systemic and pulmonary hypertension, metabolic syndrome, increased cancer risk and neuropathology (Beebe & Gozal, 2002; Drager et al. 2007; Gozal et al. 2015). CIH and/or OSA-induced neuropathologies include hippocampal cell death (Gozal et al. 2001), cognitive deficits (Engleman et al. 2000) and affective disorders (Barnes et al. 2004; Peppard et al. 2006). These morbidities are often

attributed to CIH/OSA-induced CNS inflammation (neuroinflammation) (Prabhakar et al. 2007; Gozal & Kheirandish-Gozal, 2008; Gozal, 2009; McNicholas, 2009; de Lima et al. 2016), although mechanisms of CIH-induced inflammation have scarcely been studied. In this brief review, we consider recent advances in our understanding of causes and consequences of neuroinflammation induced by multiple paradigms of hypoxia (including intermittent hypoxia and sustained hypoxia with or without rexoygenation) in the hope of gaining valuable insights concerning mechanisms of neuropathology in patients with OSA and other forms of sleep disordered breathing. Since resident CNS immune cells, microglia, are often major contributors to CNS inflammation, we focus on mechanisms regulating microglial activities, including their direct activation by hypoxia–reoxygenation (including CIH), and their indirect activation secondary to peripheral inflammation (the presence of inflammatory molecules and activated immune cells in the blood) (Gozal, 2009; Nadeem et al. 2013; Chen et al. 2015). Although certain doses of intermittent hypoxia are associated with plasticity and have functional benefits  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

(Dale et al. 2014; Navarrete-Opazo & Mitchell, 2014; Mateika et al. 2015; Peters et al. 2015), we focus on chronic intermittent hypoxia (CIH) protocols that simulate pathological aspects of sleep disordered breathing, inducing neuroinflammation and neuropathology (Dale et al. 2014; Navarrete-Opazo & Mitchell, 2014). In each section, we discuss data from OSA patients and/or experimental models of pathological CIH when available, but also present evidence from other models of hypoxia–reoxygenation and/or inflammation that may suggest neuroinflammatory mechanisms during CIH. Thus, our intent is to focus on gaps in knowledge and/or hypotheses that can be tested in the context of CIH. CIH-induced inflammation and cell death

Inflammation is a complex biological response initiated by pathogens and damaged tissue. Its primary function is to eliminate foreign bodies and repair injured tissues. A normal immune response involves the release of multiple inflammatory molecules, combined with growth/trophic (reparative) factors to heal any associated wounds. Improper control of either the inflammatory or reparative aspects of the normal response leads to chronic inflammation that exacerbates pathology (Medzhitov, 2008; Steinman, 2013). Inflammatory molecules produced include cytokines, such as tumour necrosis factor α (TNFα), interleukins (eg. IL-1β and IL-6), chemokines and adhesion molecules. The transcriptional regulation of many inflammatory cytokines, chemokines and enzymes responsible for inflammation is often controlled by pro-inflammatory transcription factors, particularly nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) (Lawrence, 2009). Molecules released from dead or dying cells (called damage-associated molecular patterns; DAMPs; Erridge, 2010) can trigger sterile inflammation (inflammation in the absence of a pathogen (Lehnardt et al. 2008)). Known DAMPs include proteins such as heat-shock proteins (e.g. HSP60) and high mobility group protein B1 (HMGB1). DAMPs may underlie immune responses observed during and following CIH. Inflammation can also be induced by production of reactive oxygen species (ROS), reactive nitrogen species and prostaglandins, produced by enzymes such as NADPH oxidase, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), respectively. These same enzymes are key players in normal inflammatory responses (Medzhitov, 2008). Rodent models of CIH and sleep disordered breathing (i.e. sleep apnoea) are associated with peripheral and CNS inflammation. OSA patients with severe sleep apnoea (greater apnoea/hypopnoea index (AHI), and/or greater oxygen desaturation independent of the AHI) exhibit increased levels of circulating adhesion molecules (Ursavas et al. 2007) as well as the cytokines TNFα and IL-6 (Ciftci  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1565

et al. 2004; Svensson et al. 2012). Similarly, rodents exposed to CIH exhibit increased NF-κB activity in cardiovascular tissues (Greenberg et al. 2006), upregulated carotid body cytokines and chemokines (Lam et al. 2012) and liver inflammation, including increased lipid peroxidation and cytokine production (Savransky et al. 2007). CNS Inflammation is also evident in rodents exosed to CIH, and it is thus predicted in OSA patients, where it is harder to establish CNS inflammation. CIH-exposed rats exhibit increased hippocampal pro-inflammatory proteins, such as heat shock proteins 60 and 70 (Gozal et al. 2002). After multiple days of CIH, rat microglia increase their expression of IL-1β, IL-6, TNFα, COX-2 and Toll-like receptor (TLR) 4 (Smith et al. 2013a), a receptor that responds to DAMPs such as HSP60 and HMGB1. CIH-induced ROS formation also increases inflammatory processes (Gao et al. 2012) and regulates pro-inflammatory enzyme expression, including iNOS and COX-2 (Zhan et al. 2005b; Block et al. 2007; Amor et al. 2010; Nair et al. 2011; Gao et al. 2012). Both iNOS and COX-2 are implicated in CIH-induced neuronal injury (Li et al. 2003, 2004) via their production of neurotoxic reactive nitrogen species and prostaglandins, respectively. Consequently, CIH increases net lipid peroxidation in the rodent CNS (Row et al. 2003). Similar ROS-dependent effects are observed in cortical neurons in vitro after even a single hypoxia–reoxygenation event; antioxidants (Burckhardt et al. 2008; Hui-guo et al. 2010) and inhibition of ROS producing enzymes (Hui-guo et al. 2010) reverse these effects, underscoring the potential for ROS to elicit CIH-induced neuronal toxicity. CIH-induced neuronal injury and death exhibit regional susceptibility. For example, there is greater injury in the cortex (Gozal et al. 2001; Row et al. 2003; Li et al. 2004; Xu et al. 2004) and hippocampus (Gozal et al. 2001; Row et al. 2003), key areas for learning and memory. CIH also promotes neural injury in CNS regions important in cardiorespiratory control such as cerebellar Purkinje neurons and the astigial nucleus (Pae et al. 2005). OSA patients and CIH-exposed rodents are reported to have reduced white matter in multiple CNS regions, suggesting demyelination and axon loss beyond neuronal apoptosis (Macey et al. 2008; Cai et al. 2012; Chen et al. 2015). Importantly, neuronal death and cognitive impairments in rodents exposed to CIH are minimized by iNOS, COX-2 and NADH oxidase inhibition (Li et al. 2003, 2004; Zhan et al. 2005a,b; Burckhardt et al. 2008; Hui-guo et al. 2010; Nair et al. 2011), suggesting that these pro-inflammatory molecules are key to CIH-induced neural injury. Peripheral inflammation, neuronal death and increased expression of pro-inflammatory molecules are consistent with a role for microglia in OSA and CIH neuropathology. In healthy brains, microglia have a dynamic, ramified morphology, involved in continuous surveillance of brain function, including synaptic activity and neuronal health,

1566

E. A. Kiernan and others

facilitating synaptic pruning or remodelling (Kettenmann et al. 2011). When cellular homeostasis is disturbed, microglia undergo complex activation processes, enabling them to migrate to the site of injury, proliferate, phagocytose dying cells or debris, and produce large quantities of neurotoxic and/or neuroprotective molecules (Kettenmann et al. 2011). Microglial activation and inflammatory molecule production contribute to neuroinflammation in almost all known neurodegenerative, traumatic and ischaemic neural disorders (Hanisch & Kettenmann, 2007). However, the specific involvement of microglia in CIH-induced neuropathology has scarcely been investigated. In the following sections, we will examine microglial activation in multiple models of hypoxia (including CIH), and explore potential mechanisms whereby CIH-induced peripheral and CNS pathology may activate microglia. Mechanisms of hypoxia-induced microglial activation: implications for CIH

Although little is known concerning mechanisms of CIH-induced neuroinflammation, even a single hypoxia–reoxygenation (HRO) event promotes inflammation in cultured microglia (Wang, 2007), and activates microglia in vivo (Smith et al. 2013b). Molecules regulating HRO-induced microglial inflammatory responses include mitogen-activated protein kinases (MAPKs), transcription factors and oxidative enzymes (Spranger et al. 1998; Vogt et al. 1998; Wang et al. 2000; Kim et al. 2003; Suk, 2004; Lai & Todd, 2006; Lu et al. 2006), many of which also play a role in peripheral CIH pathology (Yuan et al. 2011; Makarenko et al. 2014; Nanduri et al. 2015). Given the profound response of microglia to even a single HRO event, we suggest that cumulative effects from recurrent HRO may contribute to CIH-induced neuroinflammation, using many of the same molecular pathways identified for actions of CIH in peripheral tissues. Microglia may be activated by ROS from CIH per se, by factors released from injured neurons, and/or by the indirect effects of peripheral inflammation (Pineau & Lacroix, 2009). Thus, based on observations in models of hypoxia and reoxygenation, at least three mechanisms could regulate microglial activities during CIH: (1) peripheral inflammation, indirectly activating microglia via neural transmission or direct passage of pro-inflammatory molecules across the blood–brain barrier; (2) indirect microglial activation due to DAMPs released from nearby injured cells; and (3) direct microglial activation by CIH. Each potential mechanism is interrelated, and shares some common signalling mechanisms (Abstract Figure). Indirect microglial activation due to peripheral inflammation. Although the CNS is partially protected from

J Physiol 594.6

peripheral immune responses by the blood–brain barrier, peripheral inflammation nevertheless elicits CNS immune responses (Abstract Figure, A and B). Some circulating inflammatory molecules cross the blood–brain barrier into the CNS via active transport (e.g. IL-1, interleukin-1 receptor antagonist and TNFα; Wilson et al. 2002) or via circumventricular regions that lack an effective blood–brain barrier (Konsman et al. 2002). Circulating cytokines also bind to brain endothelial cells and induce proinflammatory molecules such as nitric oxide (NO) or prostaglandins that readily cross the blood–brain barrier or increase blood–brain barrier permeability (Ek et al. 2001), thus enabling CNS entry of cytokines, chemokines and peripheral immune cells. CIH may compromise the blood–brain barrier (Lim & Pack, 2014), although peripheral immune cell accumulation in the CNS of rodent CIH models has not been reported. Vagal afferent neurons may also transmit inflammatory signals to the CNS by reacting to circulating proinflammatory cytokines and rapidly inducing CNS cytokine production (Balan et al. 2011). For example, peripheral IL-1 or lipopolysaccharide (LPS; a TLR4 agonist that does not cross the blood–brain barrier) elicits brainstem inflammation within minutes, an effect abolished by bilateral vagotomy (Laye et al. 1995; Bluthe et al. 1996). Thus, increased serum levels of pro-inflammatory molecules like C-reactive protein, TNFα and IL-6 in OSA patients (Ursavas et al. 2007; Goldbart & Tal, 2008; McNicholas, 2009; Ryan et al. 2009; Svensson et al. 2012) may activate microglia. Indirect microglial activation by damage-associated molecular patterns. Microglia express TLRs, a class

of pattern recognition receptors (PRRs) activated by molecules released from stressed or dying cells (Hanisch & Kettenmann, 2007; Pineau & Lacroix, 2009). Thus, injured or stressed neurons and glia may release DAMPs into the extracellular space where they are expected to activate microglial TLRs (Pineau & Lacroix, 2009) and initiate ‘sterile’ immune responses. PRRs bind highly conserved motifs on microbial cells (pattern-associated molecular patterns; PAMPs) (Kawai & Akira, 2009). Of the four described PRR families, TLRs are most relevant in microglia; however, CNS TLRs are not uniquely expressed by microglia. TLRs are also expressed by neurons, astrocytes and cerebrovascular endothelial cells (Carty & Bowie, 2011). While TLR involvement in CIH-induced microglial activation and neuronal injury has not yet been directly investigated, CIH upregulates TLR4 in rat microglia (Smith et al. 2013a) and TLR4 contributes to OSA-induced peripheral inflammation and HRO-induced CNS inflammation (Kim et al. 2010b; Akinnusi et al. 2013). Monocytes isolated from OSA patients have elevated TLR2 and TLR4 expression (Akinnusi et al. 2013),  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

suggesting increased pro-inflammatory responses in these cells. In agreement, TLR2, TLR4 and pro-inflammatory cytokine expression in OSA patients are normalized by Continuous Positive Airway Pressure (CPAP), suggesting a causal role for CIH and/or sleep fractionation (Akinnusi et al. 2013). Further, TLR4 (Kim et al. 2010b) and TLR2/6 (Kuhlicke et al. 2007) are upregulated by hypoxia in cultured macrophages through a hypoxia-inducible transcription factor 1α (HIF-1α)-dependent pathway, potentiating LPS-induced inflammation in hypoxia (Kim et al. 2010b). DAMPs associated with OSA include circulating TLR2/TLR4 ligands, such as HMGB1, monocyte responsive protein (MRP)-8/14 (formerly S100A8/9), HSP60, fibrinogen and oxidized low density lipoprotein (Chin et al. 1996; Wessendorf et al. 2000; Saletu et al. 2006; Bhattacharjee et al. 2009; Kim et al. 2009, 2010a; Kizawa et al. 2009; Lavie et al. 2010; Wu et al. 2010; Cholidou et al. 2013). Although source(s) of DAMPs in CIH are unknown, they may mediate peripheral inflammation in OSA, and CIH-induced neuroinflammation (Lehnardt et al. 2008; Ziegler et al. 2009; Gao et al. 2011). Microglia are the CNS cells with the highest TLR expression levels, and TLR-mediated microglial inflammation is linked with neurotoxicity in neurodegenerative, infectious, traumatic and ischaemic CNS pathologies (Carty & Bowie, 2011; Okun et al. 2011; Hanamsagar et al. 2012). Microglial TLR activation by PAMPs and DAMPs elicits production of cytokines, chemokines and enzymes promoting neurotoxicity. Although TLR-mediated inflammation induces neurotoxicity with bacterial, viral, fungal and prion infection, TLR activation by endogenous ligands during sterile inflammation, such as in CIH/OSA, remains poorly understood (Okun et al. 2011). In other models of CNS injury, DAMPs released from damaged or stressed cells activate microglial TLRs (Abstract Figure, D) causing inflammation, neurotoxicity, stress or death of surrounding cells and a ‘vicious cycle’ of inflammation and cell death (Abstract Figure C; Lehnardt et al. 2003, 2008; Ziegler et al. 2009; Lehnardt, 2010). HSP60, HMGB1 and MRP8/14 are the primary DAMPs known to induce microglial inflammation (Lehnardt et al. 2008; Ziegler et al. 2009; Gao et al. 2011), and these same molecules are elevated in OSA-patient blood (Wu et al. 2010; Kim et al. 2010a) and/or the hippocampus of CIH-exposed rodents (Gozal et al. 2002). For example, in the rodent brain, CIH increases endogenous TLR4 ligands in the CNS in a region-specific manner. Proteomic analyses of hippocampal CA1 and CA3 subfields revealed increases in HSP60 and α-synuclein in the CA1 but not CA3 region of the hippocampus (Gozal et al. 2002), the primary site of hippocampal cell death (Gozal et al. 2001). Since cell death increases extracellular HSP60 release (Lehnardt et al. 2008), CIH-induced DAMPs may contribute to CA1 neuron cell death, although this remains to be tested.  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1567

Serum HMGB1 and MRP8/14 levels are increased in OSA patients (Wu et al. 2010; Kim et al. 2010a), and both have pro-inflammatory effects (Glass et al. 2010). Although HMGB1 is a nuclear protein, it is released into the extracellular space during cellular stress and inflammasome activation (Kim et al. 2006). In neurons, hypoxic/ischaemic injury leads to HMGB1 translocating from the nucleus to the cytosol where it is secreted from the cell (Faraco et al. 2007). HMGB1 activates microglial TLR2, TLR4 and/or the scavenger receptor MAC-1 (CD11b), thereby inducing inflammation (Yu et al. 2006; Gao et al. 2011). Macrophage HMGB1 release has autocrine/paracrine effects, enhancing macrophage inflammation (Kim et al. 2006; Campana et al. 2008); similar effects may also occur in microglia. MRP8/14 released from activated phagocytes enhances LPS-induced TLR4 signalling (Vogl et al. 2007) and increases MRP8/14 expression in the CNS, an indication of neuroinflammation (Vogl et al. 2007; Ziegler et al. 2009). Although HMGB1 and MRP8/14 induce TLR4-dependent inflammation following ischaemic injury (Kim et al. 2006; Faraco et al. 2007; Vogl et al. 2007; Ziegler et al. 2009), little is known concerning their roles in CIH. Since MRP8/14 oxidation reduces its pro-inflammatory activities (Lim et al. 2009), its neuroinflammatory effects on microglia may actually be constrained during CIH. Hypoxia may also directly enhance TLR4 signalling pathways, changing DAMP-induced inflammation. For example, HRO increases LPS effects on cultured microglia and potentiates iNOS, TNFα and NF-κB expression (Guo & Bhat, 2006), suggesting that HRO ‘primes’ TLR4-mediated inflammation. TLR signalling (Fig. 1) often involves recruitment of the adaptor protein myeloid differentiation factor-88 (MyD88), which initiates MAP kinase and relevant transcription factor activation, including NF-κB and HIF-1α (Hellwig-Burgel et al. 2005; Zughaier et al. 2005; Doyle & O’Neill, 2006; Lu et al. 2008). Interestingly, sustained hypoxia increases TLR4 expression in cultured microglia, and shifts TLR4 signalling from a MyD88 to a TRIF-dominant pathway (Ock et al. 2007); if a similar shift in TLR4 signalling occurs during CIH, it may attenuate CIH-induced microglial pro-inflammatory activities. Recent studies indicate that CIH increases mRNA and protein levels of TLR4 in the myocardium and the hippocampus (Yuan et al. 2014; Deng et al. 2015). It will be important to determine the hippocampal cell types in which TLR4 increases occur following CIH since cells other than microglia also express these proteins. Direct effects of CIH on microglial activities. Antioxidants

and superoxide dismutase mimetics block CIH-induced neuron death and cognitive impairment (Xu et al. 2004; Zhan et al. 2005b; Burckhardt et al. 2008; Hui-guo et al. 2010; Nair et al. 2011), but it is not yet clear if the beneficial effects of diminished ROS levels result from decreased

1568

E. A. Kiernan and others

neuronal versus microglial ROS formation. Although the direct impact of CIH on microglial ROS production has not yet been studied, it is possible that CIH directly affects microglia via ROS generation (Abstract Figure, 3). Indeed, ROS are essential for microglial inflammatory responses (Tschopp & Schroder, 2010). For example, HRO induces calcium influx through voltage-gated calcium channels and increases microglial NADPH oxidase activity, the major source of microglial extracellular ROS generation (Spranger et al. 1998). Further, HRO-induced ROS formation promotes microglial pro-inflammatory cytokine production (Spranger et al. 1998; Vogt et al. 1998; Chronic intermittent hypoxia

TLR4 DAMPs

MD2

CD14

IKKα

TRIF

IRAK2

TRAM

TRAF6 TAK1

TIRAP

IRAK4

MYD88

IKKε TAB2

TRAF3

P

IKKβ

MKKs

P

IRF3 ERK

NF-κB

p38

JNK

AP-1 Type I interferons (e.g. IFNβ), anti-inflammatory molecules

Pro-inflammatory molecules

Figure 1. Model of CIH-induced inflammation via microglial TLR4 activation Damage-associated molecular patterns (DAMPs) are endogenous TLR4 ligands that are among the factors released from stressed and dying cells. Chronic intermittent hypoxia increases several DAMPs that can bind TLR4 in microglia and activate canonical inflammatory signalling cascades. TLR4 signalling utilizes two different pathways: MyD88-dependent and TRIF-dependent. The MyD88-dependent pathway increases pro-inflammatory gene expression through activation of the MAP kinases ERK, p38 and JNK, and transcription factors including NF-κB and AP-1. The TRIF-dependent pathway induces phosphorylation of IRF3, which then translocates to the nucleus and induces expression of type I interferons (IFNs) such as IFNβ and anti-inflammatory cytokines. Both MyD88 and TRIF proteins are increased peripherally and centrally after CIH.

J Physiol 594.6

Hwang et al. 2008), and extracellular superoxide interacts synergistically with pro-inflammatory cytokines to induce neurotoxicity (Block et al. 2007). Common mechanisms of microglial activation

Mechanisms of CIH-induced microglial activation are likely to be complex; evidence from OSA patients and rodent CIH models suggests multiple activators converging on common downstream signalling pathways. In the next section, we discuss potential mechanisms of hypoxia and CIH-induced microglial activation that include (1) TLR4 activation eliciting downstream inflammasome activation; (2) MAPK activation; (3) increased expression and activity of pro-inflammatory transcription factors; and (4) possible long-lasting epigenetic modifications, including DNA methylation, hypoxia-sensitive microRNAs (miRNAs), and post-translational histone modifications. Inflammasome. The inflammasome is a high molecular mass multi-protein complex activated by ROS and TLR ligands (Lamkanfi et al. 2010; Kayagaki et al. 2011). The inflammasome complex (1) activates the enzyme caspase-1, cleaving pro-IL-1β and pro-IL-18 into mature cytokines, enabling their release through a non-canonical secretory pathway (Schroder & Tschopp, 2010) and (2) facilitates HMGB1 release (Lamkanfi et al. 2010; Kayagaki et al. 2011). Inflammasome-related molecules, including IL-18, IL-1β and HMGB1, are increased in the serum of OSA patients (Dyugovskaya et al. 2005; Minoguchi et al. 2005; Lavie & Polotsky, 2009; Wu et al. 2010). Because hypoxia upregulates caspase-11, inducing subsequent caspase-1 activity and IL-1β processing in microglial cultures (Kim et al. 2003), and HRO upregulates caspase-1 and IL-1β in the amygdala and impairs memory formation in rodents (Chiu et al. 2012), CIH may also activate the inflammasome and contribute to microglial IL-1β production (Smith et al. 2013a) and subsequent neuropathology and memory impairment (Gozal et al. 2001). Mitogen-activated protein kinases. MAPKs in immune cells upregulate both pro-inflammatory and cell survival pathway gene expression. MAPKs are activated by TLR4 ligands through a MyD88-dependent pathway (see Fig. 1), and they regulate microglial activities during sterile inflammation. Microglial p38 and ERK-1/2 MAPKs are differentially modulated by HRO. In cultured microglia, HRO activates p38, but not other MAPKs (i.e. ERK or JNK); increased p38 MAPK activity increases microglial TNFα and iNOS expression (Park et al. 2002). On the other hand, p38 MAPK inhibition blocks hypoxiaand HRO-induced iNOS activity and cytokine production (Park et al. 2002). Since HRO-induced p38 MAPK  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

activation increases caspase-11 expression, caspase-1 activation and IL-1β and IL-18 secretion (Kim et al. 2003), pro-inflammatory effects of p38 MAPK activation could be mediated in part via the inflammasome. CIH-induced inflammation in monocytes is associated with p38 MAPK activation and NF-κB activity (Ryan et al. 2007). In the cervical spinal cord, 8 h of a CIH protocol increases p38 MAPK activation in both microglia and motor neurons, and suppresses spinal phrenic motor plasticity (Huxtable et al. 2015). This effect is reversed by spinal p38 MAPK inhibition, confirming that CIH regulates microglial inflammation and its effects on neural function at least in part by MAPKs. Transcription factors. Multiple transcription factors mediate hypoxia-induced microglial inflammatory responses, including HIF-1α, NF-κB, CREB and AP-1. HIF-1α and NF-κB are well-studied and interrelated transcription factors that orchestrate complex inflammatory cascades. During normoxia, HIF-1α is hydroxylated by prolyl hydroxylases, targeting it for poly-ubiquitination, which keeps HIF-1α protein levels low (Sharp & Bernaudin, 2004; Hellwig-Burgel et al. 2005). In hypoxia, prolyl hydroxylases are inhibited, enabling HIF-1α protein accumulation, nuclear translocation and the increase of pro-inflammatory and pro-survival gene transcription. HIF-1α may also regulate innate immunity during normoxia since it is necessary for proper inflammasome function and NF-κB activation (Nizet & Johnson, 2009). TLR4 signalling activates HIF-1α independently of hypoxia, upregulating iNOS and COX-2 (Peyssonnaux et al. 2007), both of which contribute to CIH-induced neuronal death (Li et al. 2003, 2004). NF-κB, a prominent regulator of inflammatory gene transcription, is also hypoxia sensitive. During normoxia, NF-κB is sequestered in the cytosol by IκB. Activated IκB kinase-β (IKKβ) phosphorylates IκB, causing its degradation, freeing NF-κB to translocate into the nucleus (Blackwell & Christman, 1997). Prolyl hydroxylases negatively regulate IKKβ, targeting it for degradation. Thus, hypoxia permits IKKβ activation, enabling NF-κB nuclear translocation and inflammatory gene expression (Rius et al. 2008). Although sustained hypoxia preferentially increases HIF-1α (versus NF-κB) transcriptional activity, CIH preferentially increases NF-κB transcriptional activity (Ryan et al. 2005; Greenberg et al. 2006). For example, cultured monocytes from OSA patients exhibit increased NF-κB (not HIF-1α) activity in association with pro-inflammatory gene expression (Ryan et al. 2005; Greenberg et al. 2006; Htoo et al. 2006). However, these transcription factors work in concert since NF-κB is necessary for HIF-1α protein accumulation in hypoxia (Rius et al. 2008). These observations  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1569

may be cell-specific since HRO preferentially increases NF-κB activities in microglia versus astrocytes (Guo & Bhat, 2006). Although further evidence is needed, these studies suggest that CIH-induced microglial inflammatory gene expression may be at least partially regulated by NF-κB. Epigenetic regulation. Epigenetic modifications including DNA methylation, microRNAs and histone modifications underlie long-lasting and sometimes heritable changes (Portela & Esteller, 2010; Handy et al. 2011). These modifications prime cells to change transcriptional profiles in response to microenvironmental stimuli, such as hypoxia and inflammation (Muscari et al. 2013; Qiao et al. 2013; Tang et al. 2014; Cronk et al. 2015; Das et al. 2015). Epigenetic modifications may underlie long-lasting physiological or pathophysiological changes well after early life apnoeas (Bates et al. 2014) and/or CIH (Nanduri et al. 2012; Nanduri & Prabhakar, 2013; Bates et al. 2014; Prabhakar & Semenza, 2016). Although few studies directly verify that epigenetic modifications underlie long-lasting behavioural effects (i.e. breathing) in CIH or OSA, CIH-exposed neonatal rats exhibit increased DNA methylation at the superoxide dismutase 2 promoter, a modification that has lasting impact on chemoreflexes in adults (Nanduri et al. 2012). Even less is known concerning possible CIH-induced epigenetic modifications in microglia. Since hypoxia-induced microglial epigenetic modifications may regulate microglial activities long after the initiating stimulus (see below), this is an area in great need of additional research. DNA alterations may underlie epigenetic changes in peripheral cells and tissues in childhood OSA or neonatal CIH (Kim et al. 2012; Du et al. 2014; Khalyfa et al. 2014; Cortese et al. 2015), such as DNA methylation at immune/inflammatory-related genes (Kim et al. 2012). However, it is unclear if these changes persist into adulthood. Children with OSA have increased high-sensitivity C-reactive protein (hs-CRP) levels in their blood, and increased methylation of the promoter of FOXP3, a gene involved in T-cell regulation (Kim et al. 2012). Blood from children with OSA with additional abnormal postocclusive hyperaemic responses also exhibit increased DNA methylation upstream from the endothelial NOS promoter (Kheirandish-Gozal et al. 2013). Together, these observations demonstrate the potential for epigenetic modifications in peripheral immune cells following CIH, establishing a precedent for similar changes in microglia. Changes in DNA methylation regulate macrophage differentiation in tumour microenvironments (Kulshreshtha et al. 2007; Toffoli & Michiels, 2008; Garzon et al. 2010), effects that may be influenced by tumour hypoxia. This observation may help explain links between

1570

E. A. Kiernan and others

OSA and cancer where the risk of dying from cancer is higher in patients with OSA (Almendros et al. 2012; Campos-Rodriguez et al. 2013). Further research is needed to identify epigenetic mechanisms involved in CIH pathology and their relationship to cancer. There is at least one ongoing 5-year study evaluating DNA methylation and RNA transcriptome changes in the blood of OSA patients (Marin et al. 2014); results of this study will be of considerable interest. Many microRNAs (miRNAs), small non-coding RNAs that repress target gene translation, are induced by hypoxia (Nallamshetty et al. 2013). However, to date, only a few hypoxia-sensitive miRNAs have been implicated in macrophage or microglial inflammation (Taganov et al. 2006; Zhang et al. 2012; Kong et al. 2013). In primary microglia, oxygen and glucose deprivation upregulates miR-146a, miR-21, miR181a/c and miR-221 (Zhang et al. 2012; Kong et al. 2013), miRNAs with gene targets that include tumour suppressors, pro-inflammatory signalling molecules and cell cycle/survival genes. For example, miR-181c decreases TNFα production and, as a result, protects neurons from microglia-induced death (Zhang et al. 2012). Although the roles (if any) of these miRNAs in the inflammatory response to hypoxic injuries involving reoxygenation are not known, some clues may be provided by studies of miRNA regulation of TLR4 signalling, given the increase in DAMPs during CIH and OSA. The miRNAs miR-203, miR-146 and miR-155 regulate different components of TLR4 signalling (Taganov et al. 2006; Chassin et al. 2012; Qi et al. 2012; Schulte et al. 2013; Jiang et al. 2014; Yang et al. 2015). miR-203 negatively regulates microglial activation by targeting the MyD88 pathway (Yang et al. 2015), and miR-146a decreases IRAK1 and TRAF6 downstream of MyD88 in macrophages (Jiang et al. 2014). miR-155 underlies negative feedback control of macrophage TLR4 signalling (Schulte et al. 2013), and can additionally target the TRIF pathway (O’Connell et al. 2007). Thus, multiple miRNAs can constrain TLR4-induced inflammation. Whether these miRNAs or others are regulated by CIH is not yet known; however, hypoxia-dependent, epigenetic regulation of microglial inflammatory responses may occur via miRNAs. Hypoxia and pro-inflammatory molecules may also alter microglial gene transcription via histone modifications (e.g. methylation, acetylation, sumoylation and phosphorylation) (Portela & Esteller, 2010; Watson et al. 2010; Tang et al. 2014; Cho et al. 2015). The best-studied histone modification in response to hypoxia is lysine 27 methylation on histone H3 (H3K27me) via the Jumonji C (JmjC) family of histone demethylases (Shmakova et al. 2014). Unfortunately, to our knowledge, no studies of histone methylation have been done in microglia in response to any paradigm of hypoxia–reoxygenation. Although the expression of many

J Physiol 594.6

JmjC-family members is increased during hypoxia (Beyer et al. 2008; Pollard et al. 2008; Lee et al. 2014), their catalytic activity requires molecular oxygen to repress or activate specific genes (De Santa et al. 2009; Hickok et al. 2013). In macrophages, hypoxia increases H3K27 methylation (Hickok et al. 2013), indicating that hypoxia may regulate macrophage gene transcription by inhibiting this demethylase. By extrapolation from macrophages, hypoxia may have similar effects in microglia, although this remains to be directly tested, particularly in the context of CIH. Net histone methylation during hypoxia can either increase or decrease gene transcription depending on the specific methylated lysine residue. For example, histone 3 lysine 9 (H3K9) methyl marks generally repress gene transcription, whereas H3K4 methylation stimulates transcriptional activation (Bannister & Kouzarides, 2011). Hypoxia can increase both repressive (H3K9) and permissive (H3K4) methylation marks (Tausendschon et al. 2010; Zhou et al. 2010), indicating that the effects of hypoxia are likely to be gene-specific. In addition to the direct effects on histone demethylase enzyme activity via oxygen availability, hypoxia can also exert indirect effects on histone methylation. For example, NO production inhibits demethylase enzyme activity (Hickok et al. 2013); HRO increases NO production in macrophages (Park et al. 2002) and CIH increases iNOS expression in microglia (Smith et al. 2013a). Thus, histone demethylase activity may be controlled both directly and indirectly in the context of hypoxia and inflammation. Summary and concluding remarks

CIH elicits complex effects that can impact microglia in multiple ways. An understanding of the effects of CIH on microglial activities including inflammation may be essential to combat some of the neuropathology that is characteristic of sleep disordered breathing. Despite its potential importance, we know very little about mechanisms of microglial regulation in response to any profile of hypoxia, including sustained hypoxia, hypoxia–reoxygenation, therapeutic (low dose) IH and/or more severe protocols of CIH that simulate IH during sleep disordered breathing. Recent efforts to harness low-dose intermittent hypoxia as a therapeutic tool (Dale et al. 2014; Navarrete-Opazo & Mitchell, 2014) require an understanding of how intermittent hypoxia transitions from a beneficial stimulus promoting neuroplasticity without neuroinflammation, to one that promotes neuroinflammation, neuronal death and cognitive deficits. In particular, we need to know the role microglial cells play in that transition (Dale et al. 2014). Microglial activities can be both beneficial and pathological. For example, after the initial insult of  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

CIH, with associated neuronal cell death and cognitive decline, a period of reparative neurogenesis and functional recovery ensues (Gozal et al. 2001, 2003; Zhu et al. 2005, 2010; Tsai et al. 2011). We have yet to understand mechanisms underlying such compensation and partial recovery, but microglia are a known source of neurotrophic factors (e.g. brain-derived neurotrophic factor and vascular endothelial growth factor) that underlie neuroplasticity (Parkhurst & Gan, 2010), neuroprotection (Franco & Fernandez-Suarez, 2015) and neurogenesis (Sierra et al. 2014; Ribeiro Xavier et al. 2015). With greater understanding, we may optimize microglial activities to maximize reparative and protective versus deleterious and pathological effects. The CNS is a highly complex organ with extensive regional and cellular heterogeneity, and associated microenvironment differences. Indeed, microglia also exhibit regional hetergeneity (Hanisch & Kettenmann, 2007; Crain et al. 2009; Olah et al. 2011; Crain et al. 2013) that may contribute to region-specific neuronal responses when faced with intermittent hypoxia. We need to understand this heterogeneity and mechanisms that underlie microglial responses to intermittent hypoxia in short and long time domains. In this regard, of particular interest is the potential for epigenetic mechanisms to regulate microglial activities acutely and throughout life. Epigenetic regulation of microglia during intermittent hypoxia is an important area for future research. References Akinnusi M, Jaoude P, Kufel T & El-Solh AA (2013). Toll-like receptor activity in patients with obstructive sleep apnea. Sleep Breath 17, 1009–1016. Almendros I, Montserrat JM, Ramirez J, Torres M, Duran-Cantolla J, Navajas D & Farre R (2012). Intermittent hypoxia enhances cancer progression in a mouse model of sleep apnoea. Eur Respir J 39, 215–217. Amor S, Puentes F, Baker D & van der Valk P (2010). Inflammation in neurodegenerative diseases. Immunology 129, 154–169. Atalaia A, De Carvalho M, Evangelista T & Pinto A (2007). Sleep characteristics of amyotrophic lateral sclerosis in patients with preserved diaphragmatic function. Amyotroph Lateral Scler 8, 101–105. Balan KV, Kc P, Hoxha Z, Mayer CA, Wilson CG & Martin RJ (2011). Vagal afferents modulate cytokine-mediated respiratory control at the neonatal medulla oblongata. Respir Physiol Neurobiol 178, 458–464. Bannister AJ & Kouzarides T (2011). Regulation of chromatin by histone modifications. Cell Res 21, 381–395. Barnes M, McEvoy RD, Banks S, Tarquinio N, Murray CG, Vowles N & Pierce RJ (2004). Efficacy of positive airway pressure and oral appliance in mild to moderate obstructive sleep apnea. Am J Respir Crit Care Med 170, 656–664.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1571

Bates ML, Farrell ET & Eldridge MW (2014). Abnormal ventilatory responses in adults born prematurely. N Engl J Med 370, 584–585. Beebe DW & Gozal D (2002). Obstructive sleep apnea and the prefrontal cortex: towards a comprehensive model linking nocturnal upper airway obstruction to daytime cognitive and behavioral deficits. J Sleep Res 11, 1–16. Beyer S, Kristensen MM, Jensen KS, Johansen JV & Staller P (2008). The histone demethylases JMJD1A and JMJD2B are transcriptional targets of hypoxia-inducible factor HIF. J Biol Chem 283, 36542–36552. Bhattacharjee R, Kheirandish-Gozal L, Pillar G & Gozal D (2009). Cardiovascular complications of obstructive sleep apnea syndrome: evidence from children. Prog Cardiovasc Dis 51, 416–433. Blackwell TS & Christman JW (1997). The role of nuclear factor-kappa B in cytokine gene regulation. Am J Respir Cell Mol Biol 17, 3–9. Block ML, Zecca L & Hong JS (2007). Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci 8, 57–69. Bluthe RM, Michaud B, Kelley KW & Dantzer R (1996). Vagotomy attenuates behavioural effects of interleukin-1 injected peripherally but not centrally. Neuroreport 7, 1485–1488. Bombois S, Derambure P, Pasquier F & Monaca C (2010). Sleep disorders in aging and dementia. J Nutr Health Aging 14, 212–217. Burckhardt IC, Gozal D, Dayyat E, Cheng Y, Li RC, Goldbart AD & Row BW (2008). Green tea catechin polyphenols attenuate behavioral and oxidative responses to intermittent hypoxia. Am J Respir Crit Care Med 177, 1135–1141. Cai J, Tuong CM, Zhang Y, Shields CB, Guo G, Fu H & Gozal D (2012). Mouse intermittent hypoxia mimicking apnoea of prematurity: effects on myelinogenesis and axonal maturation. J Pathol 226, 495–508. Campana L, Bosurgi L & Rovere-Querini P (2008). HMGB1: a two-headed signal regulating tumor progression and immunity. Curr Opin Immunol 20, 518–523. Campos-Rodriguez F, Martinez-Garcia MA, Martinez M, Duran-Cantolla J, Pena Mde L, Masdeu MJ, Gonzalez M, Campo F, Gallego I, Marin JM, Barbe F, Montserrat JM & Farre R (2013). Association between obstructive sleep apnea and cancer incidence in a large multicenter Spanish cohort. Am J Respir Crit Care Med 187, 99–105. Carty M & Bowie AG (2011). Evaluating the role of Toll-like receptors in diseases of the central nervous system. Biochem Pharmacol 81, 825–837. Chassin C, Hempel C, Stockinger S, Dupont A, Kubler JF, Wedemeyer J, Vandewalle A & Hornef MW (2012). MicroRNA-146a-mediated downregulation of IRAK1 protects mouse and human small intestine against ischemia/reperfusion injury. EMBO Mol Med 4, 1308–1319. Chen HL, Lu CH, Lin HC, Chen PC, Chou KH, Lin WM, Tsai NW, Su YJ, Friedman M, Lin CP & Lin WC (2015). White matter damage and systemic inflammation in obstructive sleep apnea. Sleep 38, 361–370.

1572

E. A. Kiernan and others

Chin K, Ohi M, Kita H, Noguchi T, Otsuka N, Tsuboi T, Mishima M & Kuno K (1996). Effects of NCPAP therapy on fibrinogen levels in obstructive sleep apnea syndrome. Am J Respir Crit Care Med 153, 1972–1976. Chiu GS, Chatterjee D, Darmody PT, Walsh JP, Meling DD, Johnson RW & Freund GG (2012). Hypoxia/reoxygenation impairs memory formation via adenosine-dependent activation of caspase 1. J Neurosci 32, 13945–13955. Cho SH, Chen JA, Sayed F, Ward ME, Gao F, Nguyen TA, Krabbe G, Sohn PD, Lo I, Minami S, Devidze N, Zhou Y, Coppola G & Gan L (2015). SIRT1 deficiency in microglia contributes to cognitive decline in aging and neurodegeneration via epigenetic regulation of IL-1β. J Neurosci 35, 807–818. Cholidou KG, Kostakis ID, Manali ED, Perrea D, Margeli A, Vougas K, Markozannes E, Koulouris N & Alchanatis M (2013). Calprotectin: a protein related to cardiovascular risk in adult patients with obstructive sleep apnea. Cytokine 61, 917–923. Ciftci TU, Kokturk O, Bukan N & Bilgihan A (2004). The relationship between serum cytokine levels with obesity and obstructive sleep apnea syndrome. Cytokine 28, 87–91. Cortese R, Almendros I, Wang Y & Gozal D (2015). Microarray-based analysis of plasma cirDNA epigenetic modification profiling in xenografted mice exposed to intermittent hypoxia. Genomics Data 5, 17–20. Crain JM, Nikodemova M & Watters JJ (2009). Expression of P2 nucleotide receptors varies with age and sex in murine brain microglia. J Neuroinflammation 6, 24. Crain JM, Nikodemova M & Watters JJ (2013). Microglia express distinct M1 and M2 phenotypic markers in the postnatal and adult central nervous system in male and female mice. J Neurosci Res 91, 1143–1151. Cronk JC, Derecki NC, Ji E, Xu Y, Lampano AE, Smirnov I, Baker W, Norris GT, Marin I, Coddington N, Wolf Y, Turner SD, Aderem A, Klibanov AL, Harris TH, Jung S, Litvak V & Kipnis J (2015). Methyl-CpG binding protein 2 regulates microglia and macrophage gene expression in response to inflammatory stimuli. Immunity 42, 679–691. Dale EA, Ben Mabrouk F & Mitchell GS (2014). Unexpected benefits of intermittent hypoxia: enhanced respiratory and nonrespiratory motor function. Physiology (Bethesda) 29, 39–48. Das A, Chai JC, Kim SH, Lee YS, Park KS, Jung KH & Chai YG (2015). Transcriptome sequencing of microglial cells stimulated with TLR3 and TLR4 ligands. BMC Genomics 16, 517. de Lima FF, Mazzotti DR, Tufik S & Bittencourt L (2016). The role inflammatory response genes in obstructive sleep apnea syndrome: a review. Sleep Breath (in press; doi: 10.1007/s11325-015-1226-7). De Santa F, Narang V, Yap ZH, Tusi BK, Burgold T, Austenaa L, Bucci G, Caganova M, Notarbartolo S, Casola S, Testa G, Sung WK, Wei CL & Natoli G (2009). Jmjd3 contributes to the control of gene expression in LPS-activated macrophages. EMBO J 28, 3341–3352. Deng Y, Yuan X, Guo XL, Zhu D, Pan YY & Liu HG (2015). Efficacy of atorvastatin on hippocampal neuronal damage caused by chronic intermittent hypoxia: Involving TLR4 and its downstream signaling pathway. Respir Physiol Neurobiol 218, 57–63.

J Physiol 594.6

Doyle SL & O’Neill LA (2006). Toll-like receptors: from the discovery of NFkappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol 72, 1102–1113. Drager LF, Bortolotto LA, Figueiredo AC, Silva BC, Krieger EM & Lorenzi-Filho G (2007). Obstructive sleep apnea, hypertension, and their interaction on arterial stiffness and heart remodeling. Chest 131, 1379–1386. Du J, Zhang L, Zhuang S, Qin GJ & Zhao TC (2014). HDAC4 degradation mediates HDAC inhibition-induced protective effects against hypoxia/reoxygenation injury. J Cell Physiol 230, 1321–1331. Dyugovskaya L, Lavie P & Lavie L (2005). Lymphocyte activation as a possible measure of atherosclerotic risk in patients with sleep apnea. Ann N Y Acad Sci 1051, 340–350. Ek M, Engblom D, Saha S, Blomqvist A, Jakobsson PJ & Ericsson-Dahlstrand A (2001). Inflammatory response: pathway across the blood-brain barrier. Nature 410, 430–431. Engleman HM, Kingshott RN, Martin SE & Douglas NJ (2000). Cognitive function in the sleep apnea/hypopnea syndrome (SAHS). Sleep 23 Suppl 4, S102–S108. Erridge C (2010). Endogenous ligands of TLR2 and TLR4: agonists or assistants? J Leukoc Biol 87, 989–999. Faraco G, Fossati S, Bianchi ME, Patrone M, Pedrazzi M, Sparatore B, Moroni F & Chiarugi A (2007). High mobility group box 1 protein is released by neural cells upon different stresses and worsens ischemic neurodegeneration in vitro and in vivo. J Neurochem 103, 590–603. Franco R & Fernandez-Suarez D (2015). Alternatively activated microglia and macrophages in the central nervous system. Prog Neurobiol 131, 65–86. Gao HM, Zhou H & Hong JS (2012). NADPH oxidases: novel therapeutic targets for neurodegenerative diseases. Trends Pharmacol Sci 33, 295–303. Gao HM, Zhou H, Zhang F, Wilson BC, Kam W & Hong JS (2011). HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J Neurosci 31, 1081–1092. Garzon R, Marcucci G & Croce CM (2010). Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov 9, 775–789. Glass CK, Saijo K, Winner B, Marchetto MC & Gage FH (2010). Mechanisms underlying inflammation in neurodegeneration. Cell 140, 918–934. Goldbart AD & Tal A (2008). Inflammation and sleep disordered breathing in children: a state-of-the-art review. Pediatr Pulmonol 43, 1151–1160. Gozal D (2009). Sleep, sleep disorders and inflammation in children. Sleep Med 10 Suppl 1, S12–16. Gozal D, Daniel JM & Dohanich GP (2001). Behavioral and anatomical correlates of chronic episodic hypoxia during sleep in the rat. J Neurosci 21, 2442–2450. Gozal D, Farre R & Nieto FJ (2015). Putative links between sleep apnea and cancer: from hypotheses to evolving evidence. Chest 148, 1140–1147. Gozal D & Kheirandish-Gozal L (2008). Cardiovascular morbidity in obstructive sleep apnea: oxidative stress, inflammation, and much more. Am J Respir Crit Care Med 177, 369–375.  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

Gozal D, Row BW, Gozal E, Kheirandish L, Neville JJ, Brittian KR, Sachleben LR Jr & Guo SZ (2003). Temporal aspects of spatial task performance during intermittent hypoxia in the rat: evidence for neurogenesis. Eur J Neurosci 18, 2335–2342. Gozal E, Gozal D, Pierce WM, Thongboonkerd V, Scherzer JA, Sachleben LR Jr, Brittian KR, Guo SZ, Cai J & Klein JB (2002). Proteomic analysis of CA1 and CA3 regions of rat hippocampus and differential susceptibility to intermittent hypoxia. J Neurochem 83, 331–345. Greenberg H, Ye X, Wilson D, Htoo AK, Hendersen T & Liu SF (2006). Chronic intermittent hypoxia activates nuclear factor-kappaB in cardiovascular tissues in vivo. Biochem Biophys Res Commun 343, 591–596. Guo G & Bhat NR (2006). Hypoxia/reoxygenation differentially modulates NF-kappaB activation and iNOS expression in astrocytes and microglia. Antioxid Redox Signal 8, 911–918. Hanamsagar R, Hanke ML & Kielian T (2012). Toll-like receptor (TLR) and inflammasome actions in the central nervous system. Trends Immunol 33, 333–342. Handy DE, Castro R & Loscalzo J (2011). Epigenetic modifications: basic mechanisms and role in cardiovascular disease. Circulation 123, 2145–2156. Hanisch UK & Kettenmann H (2007). Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 10, 1387–1394. Hellwig-Burgel T, Stiehl DP, Wagner AE, Metzen E & Jelkmann W (2005). Review: hypoxia-inducible factor-1 (HIF-1): a novel transcription factor in immune reactions. J Interferon Cytokine Res 25, 297–310. Hickok JR, Vasudevan D, Antholine WE & Thomas DD (2013). Nitric oxide modifies global histone methylation by inhibiting Jumonji C domain-containing demethylases. J Biol Chem 288, 16004–16015. Hoch CC, Reynolds CF 3rd, Kupfer DJ, Houck PR, Berman SR & Stack JA (1986). Sleep-disordered breathing in normal and pathologic aging. J Clin Psychiatry 47, 499–503. Htoo AK, Greenberg H, Tongia S, Chen G, Henderson T, Wilson D & Liu SF (2006). Activation of nuclear factor kappaB in obstructive sleep apnea: a pathway leading to systemic inflammation. Sleep Breath 10, 43–50. Hui-guo L, Kui L, Yan-ning Z & Yong-jian X (2010). Apocynin attenuate spatial learning deficits and oxidative responses to intermittent hypoxia. Sleep Med 11, 205–212. Huxtable AG, Smith SM, Peterson TJ, Watters JJ & Mitchell GS (2015). Intermittent hypoxia-induced spinal inflammation impairs respiratory motor plasticity by a spinal p38 MAP kinase-dependent mechanism. J Neurosci 35, 6871–6880. Hwang KY, Oh YT, Yoon H, Lee J, Kim H, Choe W & Kang I (2008). Baicalein suppresses hypoxia-induced HIF-1alpha protein accumulation and activation through inhibition of reactive oxygen species and PI 3-kinase/Akt pathway in BV2 murine microglial cells. Neurosci Lett 444, 264–269. Jiang W, Kong L, Ni Q, Lu Y, Ding W, Liu G, Pu L & Tang W (2014). miR-146a ameliorates liver ischemia/reperfusion injury by suppressing IRAK1 and TRAF6. PLoS One 9, e101530. Kawai T & Akira S (2009). The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol 21, 317–337.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1573

Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, Zhang J, Lee WP, Roose-Girma M & Dixit VM (2011). Non-canonical inflammasome activation targets caspase-11. Nature 479, 117–121. Kettenmann H, Hanisch UK, Noda M & Verkhratsky A (2011). Physiology of microglia. Physiol Rev 91, 461–553. Khalyfa A, Mutskov V, Carreras A, Khalyfa AA, Hakim F & Gozal D (2014). Sleep fragmentation during late gestation induces metabolic perturbations and epigenetic changes in adiponectin gene expression in male adult offspring mice. Diabetes 63, 3230–3241. Kheirandish-Gozal L, Khalyfa A, Gozal D, Bhattacharjee R & Wang Y (2013). Endothelial dysfunction in children with obstructive sleep apnea is associated with epigenetic changes in the eNOS gene. Chest 143, 971–977. Kim J, Bhattacharjee R, Dayyat E, Snow AB, Kheirandish-Gozal L, Goldman JL, Li RC, Serpero LD, Clair HB & Gozal D (2009). Increased cellular proliferation and inflammatory cytokines in tonsils derived from children with obstructive sleep apnea. Pediatr Res 66, 423–428. Kim J, Bhattacharjee R, Khalyfa A, Kheirandish-Gozal L, Capdevila OS, Wang Y & Gozal D (2012). DNA methylation in inflammatory genes among children with obstructive sleep apnea. Am J Respir Crit Care Med 185, 330–338. Kim J, Bhattacharjee R, Snow AB, Capdevila OS, Kheirandish-Gozal L & Gozal D (2010a). Myeloid-related protein 8/14 levels in children with obstructive sleep apnoea. Eur Respir J 35, 843–850. Kim JB, Choi JS, Yu YM, Nam K, Piao CS, Kim SW, Lee MH, Han PL, Park JS & Lee JK (2006). HMGB1, a novel cytokine-like mediator linking acute neuronal death and delayed neuroinflammation in the postischemic brain. J Neurosci 26, 6413–6421. Kim NG, Lee H, Son E, Kwon OY, Park JY, Park JH, Cho GJ, Choi WS & Suk K (2003). Hypoxic induction of caspase-11/caspase-1/interleukin-1beta in brain microglia. Brain Res Mol Brain Res 114, 107–114. Kim SY, Choi YJ, Joung SM, Lee BH, Jung YS & Lee JY (2010b). Hypoxic stress up-regulates the expression of Toll-like receptor 4 in macrophages via hypoxia-inducible factor. Immunology 129, 516–524. Kizawa T, Nakamura Y, Takahashi S, Sakurai S, Yamauchi K & Inoue H (2009). Pathogenic role of angiotensin II and oxidised LDL in obstructive sleep apnoea. Eur Respir J 34, 1390–1398. Kong H, Omran A, Ashhab MU, Gan N, Peng J, He F, Wu L, Deng X & Yin F (2013). Changes in microglial inflammation-related and brain-enriched microRNAs expressions in response to in vitro oxygen-glucose deprivation. Neurochem Res 39, 233–243. Konsman JP, Parnet P & Dantzer R (2002). Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci 25, 154–159. Kuhlicke J, Frick JS, Morote-Garcia JC, Rosenberger P & Eltzschig HK (2007). Hypoxia inducible factor (HIF)-1 coordinates induction of Toll-like receptors TLR2 and TLR6 during hypoxia. PLoS One 2, e1364.

1574

E. A. Kiernan and others

Kulshreshtha R, Ferracin M, Wojcik SE, Garzon R, Alder H, Agosto-Perez FJ, Davuluri R, Liu CG, Croce CM, Negrini M, Calin GA & Ivan M (2007). A microRNA signature of hypoxia. Mol Cell Biol 27, 1859–1867. Lai AY & Todd KG (2006). Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 53, 809–816. Lam SY, Liu Y, Ng KM, Lau CF, Liong EC, Tipoe GL & Fung ML (2012). Chronic intermittent hypoxia induces local inflammation of the rat carotid body via functional upregulation of proinflammatory cytokine pathways. Histochem Cell Biol 137, 303–317. Lamkanfi M, Sarkar A, Vande Walle L, Vitari AC, Amer AO, Wewers MD, Tracey KJ, Kanneganti TD & Dixit VM (2010). Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J Immunol 185, 4385–4392. Lavie L, Dyugovskaya L, Golan-Shany O & Lavie P (2010). Heat-shock protein 70: expression in monocytes of patients with sleep apnoea and association with oxidative stress and tumour necrosis factor-alpha. J Sleep Res 19, 139–147. Lavie L & Polotsky V (2009). Cardiovascular aspects in obstructive sleep apnea syndrome–molecular issues, hypoxia and cytokine profiles. Respiration 78, 361–370. Lawrence T (2009). The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol 1, a001651. Laye S, Bluthe RM, Kent S, Combe C, Medina C, Parnet P, Kelley K & Dantzer R (1995). Subdiaphragmatic vagotomy blocks induction of IL-1 beta mRNA in mice brain in response to peripheral LPS. Am J Physiol Regul Integr Comp Physiol 268, R1327–1331. Lee HY, Choi K, Oh H, Park YK & Park H (2014). HIF-1dependent induction of Jumonji domain-containing protein (JMJD) 3 under hypoxic conditions. Mol Cells 37, 43–50. Lehnardt S (2010). Innate immunity and neuroinflammation in the CNS: the role of microglia in Toll-like receptor-mediated neuronal injury. Glia 58, 253–263. Lehnardt S, Massillon L, Follett P, Jensen FE, Ratan R, Rosenberg PA, Volpe JJ & Vartanian T (2003). Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. Proc Natl Acad Sci USA 100, 8514–8519. Lehnardt S, Schott E, Trimbuch T, Laubisch D, Krueger C, Wulczyn G, Nitsch R & Weber JR (2008). A vicious cycle involving release of heat shock protein 60 from injured cells and activation of toll-like receptor 4 mediates neurodegeneration in the CNS. J Neurosci 28, 2320–2331. Li RC, Row BW, Gozal E, Kheirandish L, Fan Q, Brittian KR, Guo SZ, Leroy R, Sachleben J & Gozal D (2003). Cyclooxygenase 2 and intermittent hypoxia-induced spatial deficits in the rat. Am J Respir Crit Care Med 168, 469–475. Li RC, Row BW, Kheirandish L, Brittian KR, Gozal E, Guo SZ, Sachleben LR Jr & Gozal D (2004). Nitric oxide synthase and intermittent hypoxia-induced spatial learning deficits in the rat. Neurobiol Dis 17, 44–53. Lim DC & Pack AI (2014). Obstructive sleep apnea and cognitive impairment: addressing the blood-brain barrier. Sleep Med Rev 18, 35–48.

J Physiol 594.6

Lim SY, Raftery MJ, Goyette J, Hsu K & Geczy CL (2009). Oxidative modifications of S100 proteins: functional regulation by redox. J Leukoc Biol 86, 577–587. Lu DY, Liou HC, Tang CH & Fu WM (2006). Hypoxia-induced iNOS expression in microglia is regulated by the PI3-kinase/Akt/mTOR signaling pathway and activation of hypoxia inducible factor-1alpha. Biochem Pharmacol 72, 992–1000. Lu YC, Yeh WC & Ohashi PS (2008). LPS/TLR4 signal transduction pathway. Cytokine 42, 145–151. Macey PM, Kumar R, Woo MA, Valladares EM, Yan-Go FL & Harper RM (2008). Brain structural changes in obstructive sleep apnea. Sleep 31, 967–977. Makarenko VV, Usatyuk PV, Yuan G, Lee MM, Nanduri J, Natarajan V, Kumar GK & Prabhakar NR (2014). Intermittent hypoxia-induced endothelial barrier dysfunction requires ROS-dependent MAP kinase activation. Am J Physiol Cell Physiol 306, C745–752. Manon-Espaillat R, Gothe B, Ruff RL & Newman C (1989). Sleep apnea in multiple sclerosis. Neurorehabil Neural Repair 3, 133–136. Marcus CL, Keens TG, Bautista DB, von Pechmann WS & Ward SL (1991). Obstructive sleep apnea in children with Down syndrome. Pediatrics 88, 132–139. Marin JM, Artal J, Martin T, Carrizo SJ, Andres M, Martin-Burriel I, Bolea R, Sanz A, Varona L, Godino J, Gallego B, Garcia-Erce JA, Villar I, Gil V, Forner M, Cubero JP & Ros L (2014). Epigenetics modifications and subclinical atherosclerosis in obstructive sleep apnea: The EPIOSA study. BMC Pulm Med 14, 114. Mateika JH, El-Chami M, Shaheen D & Ivers B (2015). Intermittent hypoxia: a low-risk research tool with therapeutic value in humans. J Appl Physiol (1985) 118, 520–532. McNicholas WT (2009). Obstructive sleep apnea and inflammation. Prog Cardiovasc Dis 51, 392–399. Medzhitov R (2008). Origin and physiological roles of inflammation. Nature 454, 428–435. Minoguchi K, Yokoe T, Tazaki T, Minoguchi H, Tanaka A, Oda N, Okada S, Ohta S, Naito H & Adachi M (2005). Increased carotid intima-media thickness and serum inflammatory markers in obstructive sleep apnea. Am J Respir Crit Care Med 172, 625–630. Muscari C, Giordano E, Bonafe F, Govoni M, Pasini A & Guarnieri C (2013). Priming adult stem cells by hypoxic pretreatments for applications in regenerative medicine. J Biomed Sci 20, 63. Nadeem R, Molnar J, Madbouly EM, Nida M, Aggarwal S, Sajid H, Naseem J & Loomba R (2013). Serum inflammatory markers in obstructive sleep apnea: a meta-analysis. J Clin Sleep Med 9, 1003–1012. Nair D, Dayyat EA, Zhang SX, Wang Y & Gozal D (2011). Intermittent hypoxia-induced cognitive deficits are mediated by NADPH oxidase activity in a murine model of sleep apnea. PLoS One 6, e19847. Nallamshetty S, Chan SY & Loscalzo J (2013). Hypoxia: a master regulator of microRNA biogenesis and activity. Free Radic Biol Med 64, 20–30.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

Nanduri J, Makarenko V, Reddy VD, Yuan G, Pawar A, Wang N, Khan SA, Zhang X, Kinsman B, Peng YJ, Kumar GK, Fox AP, Godley LA, Semenza GL & Prabhakar NR (2012). Epigenetic regulation of hypoxic sensing disrupts cardiorespiratory homeostasis. Proc Natl Acad Sci USA 109, 2515–2520. Nanduri J, Peng YJ, Yuan G, Kumar GK & Prabhakar NR (2015). Hypoxia-inducible factors and hypertension: lessons from sleep apnea syndrome. J Mol Med (Berl) 93, 473–480. Nanduri J & Prabhakar NR (2013). Developmental programming of O2 sensing by neonatal intermittent hypoxia via epigenetic mechanisms. Respir Physiol Neurobiol 185, 105–109. Navarrete-Opazo A & Mitchell GS (2014). Therapeutic potential of intermittent hypoxia: a matter of dose. Am J Physiol Regul Integr Comp Physiol 307, R1181–R1197. Nizet V & Johnson RS (2009). Interdependence of hypoxic and innate immune responses. Nat Rev Immunol 9, 609–617. O’Connell RM, Taganov KD, Boldin MP, Cheng G & Baltimore D (2007). MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA 104, 1604–1609. Ock J, Jeong J, Choi WS, Lee WH, Kim SH, Kim IK & Suk K (2007). Regulation of Toll-like receptor 4 expression and its signaling by hypoxia in cultured microglia. J Neurosci Res 85, 1989–1995. Okun E, Griffioen KJ & Mattson MP (2011). Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci 34, 269–281. Olah M, Biber K, Vinet J & Boddeke HW (2011). Microglia phenotype diversity. CNS Neurol Disord Drug Targets 10, 108–118. Pae EK, Chien P & Harper RM (2005). Intermittent hypoxia damages cerebellar cortex and deep nuclei. Neurosci Lett 375, 123–128. Park JG, Ramar K & Olson EJ (2011). Updates on definition, consequences, and management of obstructive sleep apnea. Mayo Clin Proc 86, 549–554; quiz 554–545. Park SY, Lee H, Hur J, Kim SY, Kim H, Park JH, Cha S, Kang SS, Cho GJ, Choi WS & Suk K (2002). Hypoxia induces nitric oxide production in mouse microglia via p38 mitogen-activated protein kinase pathway. Brain Res Mol Brain Res 107, 9–16. Parkhurst CN & Gan WB (2010). Microglia dynamics and function in the CNS. Curr Opin Neurobiol 20, 595–600. Peppard PE, Szklo-Coxe M, Hla KM & Young T (2006). Longitudinal association of sleep-related breathing disorder and depression. Arch Intern Med 166, 1709–1715. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW & Hla KM (2013). Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol 177, 1006–1014. Peters ME, Kimyon RS, Mitchell GS & Watters JJ (2015). Repetitive acute intermittent hypoxia does not promote generalized inflammatory gene expression in the rat CNS. Respir Physiol Neurobiol 218, 1–10. Peyssonnaux C, Cejudo-Martin P, Doedens A, Zinkernagel AS, Johnson RS & Nizet V (2007). Cutting edge: Essential role of hypoxia inducible factor-1alpha in development of lipopolysaccharide-induced sepsis. J Immunol 178, 7516–7519.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1575

Pineau I & Lacroix S (2009). Endogenous signals initiating inflammation in the injured nervous system. Glia 57, 351–361. Pollard PJ, Loenarz C, Mole DR, McDonough MA, Gleadle JM, Schofield CJ & Ratcliffe PJ (2008). Regulation of Jumonji-domain-containing histone demethylases by hypoxia-inducible factor (HIF)-1alpha. Biochem J 416, 387–394. Portela A & Esteller M (2010). Epigenetic modifications and human disease. Nat Biotechnol 28, 1057–1068. Prabhakar NR, Kumar GK, Nanduri J & Semenza GL (2007). ROS signaling in systemic and cellular responses to chronic intermittent hypoxia. Antioxid Redox Signal 9, 1397–1403. Prabhakar NR & Semenza GL (2016). Regulation of carotid body oxygen sensing by hypoxia-inducible factors. Pflugers Arch 481, 71–75. Qi J, Qiao Y, Wang P, Li S, Zhao W & Gao C (2012). microRNA-210 negatively regulates LPS-induced production of proinflammatory cytokines by targeting NF-kappaB1 in murine macrophages. FEBS Lett 586, 1201–1207. Qiao Y, Giannopoulou EG, Chan CH, Park SH, Gong S, Chen J, Hu X, Elemento O & Ivashkiv LB (2013). Synergistic activation of inflammatory cytokine genes by interferon-gamma-induced chromatin remodeling and toll-like receptor signaling. Immunity 39, 454–469. Ribeiro Xavier AL, Kress BT, Goldman SA, Lacerda de Menezes JR & Nedergaard M (2015). A distinct population of microglia supports adult neurogenesis in the subventricular zone. J Neurosci 35, 11848–11861. Rius J, Guma M, Schachtrup C, Akassoglou K, Zinkernagel AS, Nizet V, Johnson RS, Haddad GG & Karin M (2008). NF-κB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1α. Nature 453, 807–811. Row BW, Liu R, Xu W, Kheirandish L & Gozal D (2003). Intermittent hypoxia is associated with oxidative stress and spatial learning deficits in the rat. Am J Respir Crit Care Med 167, 1548–1553. Ryan S, McNicholas WT & Taylor CT (2007). A critical role for p38 map kinase in NF-kappaB signaling during intermittent hypoxia/reoxygenation. Biochem Biophys Res Commun 355, 728–733. Ryan S, Taylor CT & McNicholas WT (2005). Selective activation of inflammatory pathways by intermittent hypoxia in obstructive sleep apnea syndrome. Circulation 112, 2660–2667. Ryan S, Taylor CT & McNicholas WT (2009). Systemic inflammation: a key factor in the pathogenesis of cardiovascular complications in obstructive sleep apnoea syndrome? Thorax 64, 631–636. Saletu M, Nosiska D, Kapfhammer G, Lalouschek W, Saletu B, Benesch T & Zeitlhofer J (2006). Structural and serum surrogate markers of cerebrovascular disease in obstructive sleep apnea (OSA): association of mild OSA with early atherosclerosis. J Neurol 253, 746–752. Savransky V, Bevans S, Nanayakkara A, Li J, Smith PL, Torbenson MS & Polotsky VY (2007). Chronic intermittent hypoxia causes hepatitis in a mouse model of diet-induced fatty liver. Am J Physiol Gastrointest Liver Physiol 293, G871–G877.

1576

E. A. Kiernan and others

Schroder K & Tschopp J (2010). The inflammasomes. Cell 140, 821–832. Schulte LN, Westermann AJ & Vogel J (2013). Differential activation and functional specialization of miR-146 and miR-155 in innate immune sensing. Nucleic Acids Res 41, 542–553. Sharp FR & Bernaudin M (2004). HIF1 and oxygen sensing in the brain. Nat Rev Neurosci 5, 437–448. Shmakova A, Batie M, Druker J & Rocha S (2014). Chromatin and oxygen sensing in the context of JmjC histone demethylases. Biochem J 462, 385–395. Sierra A, Beccari S, Diaz-Aparicio I, Encinas JM, Comeau S & Tremblay ME (2014). Surveillance, phagocytosis, and inflammation: how never-resting microglia influence adult hippocampal neurogenesis. Neural Plast 2014, 610343. Smith SM, Friedle SA & Watters JJ (2013a). Chronic intermittent hypoxia exerts CNS region-specific effects on rat microglial inflammatory and TLR4 gene expression. PLoS One 8, e81584. Smith SM, Mitchell GS, Friedle SA, Sibigtroth CM, Vinit S & Watters JJ (2013b). Hypoxia attenuates purinergic P2X receptor-induced inflammatory gene expression in brainstem microglia. Hypoxia (Auckl) 2013, doi: 10.2147/HP.S45529. Spranger M, Kiprianova I, Krempien S & Schwab S (1998). Reoxygenation increases the release of reactive oxygen intermediates in murine microglia. J Cereb Blood Flow Metab 18, 670–674. Steinman L (2013). Inflammatory cytokines at the summits of pathological signal cascades in brain diseases. Sci Signal 6, pe3. Suk K (2004). Minocycline suppresses hypoxic activation of rodent microglia in culture. Neurosci Lett 366, 167–171. Svensson M, Venge P, Janson C & Lindberg E (2012). Relationship between sleep-disordered breathing and markers of systemic inflammation in women from the general population. J Sleep Res 21, 147–154. Taganov KD, Boldin MP, Chang KJ & Baltimore D (2006). NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA 103, 12481–12486. Tang Y, Li T, Li J, Yang J, Liu H, Zhang XJ & Le W (2014). Jmjd3 is essential for the epigenetic modulation of microglia phenotypes in the immune pathogenesis of Parkinson’s disease. Cell Death Differ 21, 369–380. Tausendschon M, Dehne N & Brune B (2010). Hypoxia causes epigenetic gene regulation in macrophages by attenuating Jumonji histone demethylase activity. Cytokine 53, 256–262. Toffoli S & Michiels C (2008). Intermittent hypoxia is a key regulator of cancer cell and endothelial cell interplay in tumours. FEBS J 275, 2991–3002. Tran K, Hukins C, Geraghty T, Eckert B & Fraser L (2010). Sleep-disordered breathing in spinal cord-injured patients: a short-term longitudinal study. Respirology 15, 272–276. Tsai YW, Yang YR, Wang PS & Wang RY (2011). Intermittent hypoxia after transient focal ischemia induces hippocampal neurogenesis and c-Fos expression and reverses spatial memory deficits in rats. PLoS One 6, e24001.

J Physiol 594.6

Tschopp J & Schroder K (2010). NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat Rev Immunol 10, 210–215. Ursavas A, Karadag M, Rodoplu E, Yilmaztepe A, Oral HB & Gozu RO (2007). Circulating ICAM-1 and VCAM-1 levels in patients with obstructive sleep apnea syndrome. Respiration 74, 525–532. Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA, Nacken W, Foell D, van der Poll T, Sorg C & Roth J (2007). Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med 13, 1042–1049. Vogt M, Bauer MK, Ferrari D & Schulze-Osthoff K (1998). Oxidative stress and hypoxia/reoxygenation trigger CD95 (APO-1/Fas) ligand expression in microglial cells. FEBS Lett 429, 67–72. Wang JY (2007). Hypoxia/reoxygenation induces nitric oxide and TNF-alpha release from cultured microglia but not astrocytes of the rat. Chin J Physiol 50, 127–134. Wang JY, Shum AY, Chao CC & Kuo JS (2000). Production of macrophage inflammatory protein-2 following hypoxia/reoxygenation in glial cells. Glia 32, 155–164. Watson JA, Watson CJ, McCann A & Baugh J (2010). Epigenetics, the epicenter of the hypoxic response. Epigenetics 5, 293–296. Wessendorf TE, Thilmann AF, Wang YM, Schreiber A, Konietzko N & Teschler H (2000). Fibrinogen levels and obstructive sleep apnea in ischemic stroke. Am J Respir Crit Care Med 162, 2039–2042. Wilson CJ, Finch CE & Cohen HJ (2002). Cytokines and cognition–the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc 50, 2041–2056. Wu KM, Lin CC, Chiu CH & Liaw SF (2010). Effect of treatment by nasal continuous positive airway pressure on serum high mobility group box-1 protein in obstructive sleep apnea. Chest 137, 303–309. Xu W, Chi L, Row BW, Xu R, Ke Y, Xu B, Luo C, Kheirandish L, Gozal D & Liu R (2004). Increased oxidative stress is associated with chronic intermittent hypoxia-mediated brain cortical neuronal cell apoptosis in a mouse model of sleep apnea. Neuroscience 126, 313–323. Yang Z, Zhong L, Zhong S, Xian R & Yuan B (2015). miR-203 protects microglia mediated brain injury by regulating inflammatory responses via feedback to MyD88 in ischemia. Mol Immunol 65, 293–301. Yu M, Wang H, Ding A, Golenbock DT, Latz E, Czura CJ, Fenton MJ, Tracey KJ & Yang H (2006). HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock 26, 174–179. Yuan G, Khan SA, Luo W, Nanduri J, Semenza GL & Prabhakar NR (2011). Hypoxia-inducible factor 1 mediates increased expression of NADPH oxidase-2 in response to intermittent hypoxia. J Cell Physiol 226, 2925–2933. Yuan X, Deng Y, Guo X, Shang J, Zhu D & Liu H (2014). Atorvastatin attenuates myocardial remodeling induced by chronic intermittent hypoxia in rats: partly involvement of TLR-4/MYD88 pathway. Biochem Biophys Res Commun 446, 292–297.  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.6

Mechanisms of microglial activation in chronic intermittent hypoxia

Zhan G, Fenik P, Pratico D & Veasey SC (2005a). Inducible nitric oxide synthase in long-term intermittent hypoxia: hypersomnolence and brain injury. Am J Respir Crit Care Med 171, 1414–1420. Zhan G, Serrano F, Fenik P, Hsu R, Kong L, Pratico D, Klann E & Veasey SC (2005b). NADPH oxidase mediates hypersomnolence and brain oxidative injury in a murine model of sleep apnea. Am J Respir Crit Care Med 172, 921–929. Zhang L, Dong L-Y, Li Y-J, Hong Z & Wei W-S (2012). The microRNA miR-181c controls microglia-mediated neuronal apoptosis by suppressing tumor necrosis factor. J Neuroinflammation 9, 211. Zhou X, Sun H, Chen H, Zavadil J, Kluz T, Arita A & Costa M (2010). Hypoxia induces trimethylated H3 lysine 4 by inhibition of JARID1A demethylase. Cancer Res 70, 4214–4221. Zhu LL, Zhao T, Li HS, Zhao H, Wu LY, Ding AS, Fan WH & Fan M (2005). Neurogenesis in the adult rat brain after intermittent hypoxia. Brain Res 1055, 1–6. Zhu XH, Yan HC, Zhang J, Qu HD, Qiu XS, Chen L, Li SJ, Cao X, Bean JC, Chen LH, Qin XH, Liu JH, Bai XC, Mei L & Gao TM (2010). Intermittent hypoxia promotes hippocampal neurogenesis and produces antidepressant-like effects in adult rats. J Neurosci 30, 12653–12663.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

1577

Ziegler G, Prinz V, Albrecht MW, Harhausen D, Khojasteh U, Nacken W, Endres M, Dirnagl U, Nietfeld W & Trendelenburg G (2009). Mrp-8 and -14 mediate CNS injury in focal cerebral ischemia. Biochim Biophys Acta 1792, 1198–1204. Zughaier SM, Zimmer SM, Datta A, Carlson RW & Stephens DS (2005). Differential induction of the toll-like receptor 4-MyD88-dependent and -independent signaling pathways by endotoxins. Infect Immun 73, 2940–2950.

Additional information Competing interests None declared. Author contributions All authors have approved the final version of the manuscript and agree to be accountable for all aspects of the work. All persons designated as authors qualify for authorship, and all those who qualify for authorship are listed. Funding Supported by: NIH training grants GM0075007 (EAK) and HL07694 (SMCS), and NIH R01 HL111598 (GSM and JJW).

Mechanisms of microglial activation in models of inflammation and hypoxia: Implications for chronic intermittent hypoxia.

Chronic intermittent hypoxia (CIH) is a hallmark of sleep apnoea, a condition associated with diverse clinical disorders. CIH and sleep apnoea are cha...
492KB Sizes 0 Downloads 8 Views