Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Apr 1.
Published in final edited form as: Prog Neurobiol. 2017 Oct 12;163-164:27–58. doi: 10.1016/j.pneurobio.2017.10.002

Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair

Wei Cai a,1, Tuo Yang a,1, Huan Liu a, Lijuan Han a, Kai Zhang a, Xiaoming Hu a,b,e, Xuejing Zhang a, Ke-Jie Yin a, Yanqin Gao b, Michael V L Bennett c, Rehana K Leak d, Jun Chen a,b,e
PMCID: PMC6037317  NIHMSID: NIHMS940339  PMID: 29032144

Abstract

Peroxisome proliferator-activated receptor γ (PPARγ) is a widely expressed ligand-modulated transcription factor that governs the expression of genes involved in inflammation, redox equilibrium, trophic factor production, insulin sensitivity, and the metabolism of lipids and glucose. Synthetic PPARγ agonists (e.g. thiazolidinediones) are used to treat Type II diabetes and have the potential to limit the risk of developing brain injuries, such as stroke, by mitigating the influence of comorbidities. If brain injury develops, PPARγ serves as a master gatekeeper of cytoprotective stress responses, improving the chances of cellular survival and recovery of homeostatic equilibrium. In the acute injury phase, PPARγ directly restricts tissue damage by inhibiting the NFκB pathway to mitigate inflammation and stimulating the Nrf2/ARE axis to neutralize oxidative stress. During the chronic phase of acute brain injuries, PPARγ activation in injured cells culminates in the repair of gray and white matter, preservation of the blood-brain barrier, reconstruction of the neurovascular unit, resolution of inflammation, and long-term functional recovery. Thus, PPARγ lies at the apex of cell fate decisions and exerts profound effects on the chronic progression of acute injury conditions. Here, we review the therapeutic potential of PPARγ in stroke and brain trauma and highlight the novel role of PPARγ in long-term tissue repair. We describe its structure and function and identify the genes that it targets. PPARγ regulation of inflammation, metabolism, cell fate (proliferation/differentiation/maturation/survival), and many other processes also has relevance to other neurological diseases. Therefore, PPARγ is an attractive target for therapies against a number of progressive neurological disorders.

Keywords: Thiazolidinedione, Nrf2, stroke, traumatic brain injury, inflammation, remyelination

1. Introduction

Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the pleiotropic nuclear receptor 1C (NR1C) family (also known as the PPAR family) (Janani and Ranjitha Kumari, 2015). This nuclear receptor family encompasses a group of ligand-modulated transcription factors with broad tissue distributions and a wide array of target genes and functions (Grygiel-Gorniak, 2014). PPARγ is a master gatekeeper of the expression of numerous genes, including G-protein coupled receptors, growth factors, antioxidant enzymes, stem cell genes, kinases, cytokines/chemokines, pro-inflammatory factors, ion channels, and transporters. PPARγ plays a well-established role in the regulation of adipocyte differentiation and the metabolism of lipid and glucose (Janani and Ranjitha Kumari, 2015). Thus, synthetic PPARγ ligands such as thiazolidinediones (TZDs or glitazones) are currently prescibed worldwide to treat hyperglycemia and diabetes and can be readily side-tracked for use in other conditions. Aside from its role in metabolic regulation, anti-inflammatory and protective effects of PPARγ have also been widely studied. For example, PPARγ activation is known to mitigate neuroinflammation and exert direct neuronal protection after central nervous system (CNS) injuries (Gillespie et al., 2011).

Acute CNS injuries are among the leading causes of disability, mortality, and morbidity worldwide (Murray and Lopez, 1997). Tissue damage in these conditions is elicited by both primary and secondary mechanisms, and impacts almost every component of the CNS, including grey matter, white matter, and the vascular network. A growing body of evidence indicates that PPARγ is critically involved in the long-term promotion of tissue repair and rescue of brain cells. In addition, PPARγ directly or indirectly controls the manifestation of comorbid diseases such as diabetes and hypertension, which can profoundly influence the onset and outcome of CNS injuries. Given its dominant position as a master gatekeeper of gene expression, PPARγ is an attractive therapeutic target for injury conditions such as ischemic stroke, hemorrhagic stroke, traumatic brain injury (TBI), and spinal cord injury (SCI).

In this article, we briefly review the numerous functions of PPARγ and their effects on CNS injury. We begin by describing the pathophysiological mechanisms underlying CNS injury in both the acute injury phase and the chronic repair phase. We discuss the contributions of comorbidities to increased disease incidence and negative clinical outcomes and their modulation by PPARγ. The key role of PPARγ in tissue repair and regeneration is highlighted throughout this review. Finally, we will end with our perspectives on PPARγ-related translational therapies.

2. PPARγ overview

2.1 PPARγ structure and CNS distribution

PPARs, or NR1Cs, were first cloned in rodent hepatocytes in the 1990s, and found to be activated by a group of peroxisome proliferators (Issemann and Green, 1990). PPARs are ligand-activated transcription factors that influence the expression of a number of genes, among which metabolism-related genes are the best documented (Michalik and Wahli, 2006). Three isotypes of PPARs have been identified in mammals—PPARα (NR1C1), δ/β (NR1C2), and γ (NR1C3). As with all other nuclear receptors, PPARs share similar structural features and contain the following functional domains: a N-terminal ligand-independent functional domain, a DNA-binding domain that specifically recognizes peroxisome proliferator response elements (PPREs), a flexible hinge region, and a C-terminal ligand binding domain (LBD) (Zoete et al., 2007). The three PPAR isotypes are encoded by genes located on different chromosomes (Greene et al., 1995) and exhibit distinct tissue distribution patterns and biological functions. PPARα is mainly expressed in tissues with high fatty acid metabolism, including liver, kidney, and white and brown adipose tissue (Delerive et al., 2000; Lo Verme et al., 2005; Neschen et al., 2007). PPARδ/β is ubiquitously and abundantly expressed in a broad range of tissues and regulates fatty acid oxidation (Wang et al., 2003b; Stephen et al., 2004). PPARγ consists of two isoforms in human and rodents—PPARγ1, a ~54.5 kDa shorter form, and PPARγ2, a ~57.6 kDa longer form (Chen et al., 2012) (Figure 1A). PPARγ2 is restricted to adipose tissue and plays an important role in adipocyte differentiation. In contrast, PPARγ1 is expressed in various cell types, including brain cells such as neurons and glia, and bone marrow derived immune cells (Zhu et al., 1995; Elbrecht et al., 1996)

Figure 1.

Figure 1

PPARγ structure and post-translational modifications (PTMs). (A) Human PPARγ consists of two isoforms, PPARγ1 (477 aa) and PPARγ2 (505 aa). Like all other nuclear receptors, PPARγ has four functional domains: a ligand-independent activation domain, a DNA binding domain, a hinge domain, and a ligand-binding domain. (B) PPARγ activity is regulated by PTMs. Ser112 phosphorylation results in different transcriptional outcomes depending on the kinases involved. CDK7/9 mediated Ser112 phosphorylation leads to increased PPARγ activity whereas MAPK elicits the opposite effect. Lys107 SUMOylation is associated with increases in PPARγ activity. Ser273 phosphorylation results in decreased insulin sensitivity. SUMOylation of Lys395 mediates transrepression. Abbreviations: Mitogen-activated protein kinases (MAPKs), cyclin-dependent kinase (CDK).

Post-translational modulations (PTMs) of specific amino acids influence PPARγ activity and its functional states (Kim et al., 2013). The major types of PTMs that regulate PPARγ function are phosphorylation, SUMOylation, and ubiquitination (Figure 1B). Ser112 phosphorylation decreases PPARγ activity if induced by mitogen-activated protein kinases (Hu et al., 1996; Shao et al., 1998), but increases PPARγ activity if induced by cyclin-dependent kinase (CDK) 7 and CDK9 (Compe et al., 2005; Iankova et al., 2006). CDK5-mediated Ser273 phosphorylation of PPARγ leads to reduced insulin sensitivity (Choi et al., 2010; Choi et al., 2011). SUMOylation at Lys107 results in decreased activity, whereas Lys395 SUMOylation is strongly associated with PPARγ transrepression of nuclear factor (NF)-κB (Yamashita et al., 2004; Pascual et al., 2005; Jennewein et al., 2008; Pourcet et al., 2010). The latter function is particularly important in anti-inflammatory effects and will be descibed in Section 2.3. Ubiquitination of PPARγ tags it for proteasomal degradation, a process that is enhanced by interferon (IFN)-γ (Waite et al., 2001) and tumor necrosis factor (TNF)-α (He et al., 2008), and repressed by sirtuin 1 (Picard et al., 2004; Floyd et al., 2008).

All three subtypes of PPARs are expressed in the CNS, albeit at different levels (Elbrecht et al., 1996; Braissant and Wahli, 1998; Cullingford et al., 1998; Moreno et al., 2004). Whereas PPAR δ/β is broadly and robustly expressed in the CNS, PPARα and PPARγ exhibit a more restricted pattern of distribution (Braissant and Wahli, 1998; Cullingford et al., 1998; Moreno et al., 2004). Under physiological conditions, PPARγ is readily detectable in select areas, such as the basal ganglia, thalamus, piriform cortex, and hippocampus (Braissant and Wahli, 1998 ; Moreno et al., 2004) and this expression is mainly in neuronal cells (Ferguson et al., 2014). A fraction of astrocytes (20-40%) also express PPARγ, predominantly in processes rather than somata (Warden et al., 2016). Although PPARγ expression has been observed in microglial cultures (Bernardo et al., 2000), it is barely detectable in this cell type in vivo under physiological conditions. However, lipopolysaccharide (LPS) stiumlation significantly increases microglial PPARγ expression in the brain, indicating that microglial PPARγ expression may be dependent on inflammation status (Warden et al., 2016).

2.2 PPARγ ligands

PPARγ ligands can be classified into three major categories. Category A includes natural (endogenous) agonists, such as unsaturated fatty acids, eicosnanoids, oxidized phospholipids, and nitroalkenes. Among these agonists, an eicosanoid termed prostaglandin metabolite 15-deoxy-D12, 14 prostaglandin J2 (15d-PGJ2) was the first specific endogenous ligand of PPARγ to be identified and has been widely used in studies of PPARγ in various disease models (Forman et al., 1995). Category B includes synthetic agonists: 1) TZDs such as pioglitazone (PGZ), ciglitazone (CGZ), triglitazone (TGZ), and rosiglitazone (RGZ), 2) non-TZD agonists, 3) dual-α/γ agonists, 4) pan-α/β/γ agonists, and 5) selective PPARγ modulators. Among these, TZDs were the first to be synthesized and are widely used in diabetic patients (Lehmann et al., 1995), as well as in experimental models to assess the effects of PPARγ activation. Category C includes synthetic antagonists, such as bisphenol A diglyceryl ether, GW9662, LG100641, PD068235, T0070907, and SR-202. Among these, GW9662 is an irreversible PPARγ antagonist that has been widely applied in research on PPARγ (Leesnitzer et al., 2002; Chen et al., 2012).

It is important to note that almost all of the aforementioned PPARγ agonists not only activate PPARγ, but also activate other PPARs, as well as PPAR-independent pathways (Park et al., 2003; Bernardo et al., 2009). As a result, the effects of PPARγ agonists, including TZDs, work through both PPARγ-dependent and independent mechanisms that are not easily differentiated and can confound the interpretations of studies. Furthermore, regardless of the route of delivery (iv, ip, oral, or intracerebral), TZDs can affect PPARγ in the CNS (Culman et al., 2007), although it is not known whether all TZDs cross the blood-brain barrier (BBB). PGZ has been reported to cross the BBB (Grommes et al., 2013), but unpublished observations by GlaxoSmithKline have been used to claim that RGZ cannot cross the BBB (Pedersen et al., 2006), although this awaits verification.

2.3 PPARγ functioning patterns

As with all other nuclear receptors, PPARγ functioning patterns involve ligand-independent repression, ligand-dependent transactivation, and ligand-dependent transrepression (Glass and Ogawa, 2006) (Figure 2). As there are no known endogeous PPARγ-inhibiting ligands, all “ligands” in this review refer to PPARγ-activating ligands.

Figure 2.

Figure 2

Functional patterns of PPARγ. (A) Ligand-independent repression and ligand-dependent transactivation. Under resting conditions, the PPARγ/RXR heterodimer is connected to co-repressor (HDAC and NCoR), which blocks PPRE target gene transcription. After binding to the PPARy ligand, conformational changes lead to dissociation of the co-repressor and recruitment of a co-activator (e.g. TRAP220), which triggers transcription of PPRE target genes. (B) Ligand-dependent transrepression. Binding to its ligand leads to SUMOylation of PPARγ, which stabilizes the co-repressor of NF-κB, leading to blockade of NF-κB target gene expression. (C) PPARγ can also inhibit NF-κB target genes by 1) mediating NF-κB degradation, 2) facilitating NF-κB export out of the nucleus, 3) cofactor competition with NF-κB, and 4) steric inhibition of NF-κB binding. Abbreviations: retinoid X receptor (RXR), histone deacetylase (HCAC), nuclear receptor corepressor (NCoR), thyroid hormone-associated protein 220 (TRAP220), peroxisome proliferator response elements (PPREs), ubiquitin (Ub), nuclear factor-κB (NF-κB).

The classic view of PPARγ function is that it forms a heterodimer with the retinoid X receptor (RXR) (Berger and Moller, 2002). Under basal conditions, in the absence of ligands of RXR or PPARγ, the heterodimer is associated with corepressors, such as nuclear receptor corepressor (NCoR), transducin β-like protein, and histone deacetylases (HDACs). This complex binds to PPREs in the promoter region of target genes and retains the genes in a suppressed state defined as ligand-independent repression. Following ligand binding, however, the PPARγ/RXR heterodimer undergoes conformational changes, dissociates from co-repressors, and recruits co-activators such as thyroid hormone-associated protein 220, thereby upregulating PPRE target gene transcription (Dekkers et al., 2012). This effect is known as ligand-dependent transactivation (Figure 2A). Ligands of either PPARγ or RXR can both initiate ligand-dependent transactivation, whereas binding of both receptors simultaneously can lead to additive effects on gene expression (Fajas et al., 1997). Both ligand-independent repression and ligand-dependent transactivation involve the regulation of promoters located at PPREs, and PPARγ is known to maintain lipid metabolic homeostasis by these mechanisms.

Ligand-dependent transrepression is more complex and denotes PPARγ-dependent repression of genes other than PPREs, typically NF-κB and activator protein-1, in the absence of direct binding of PPARγ to DNA. The HDAC3/NCoR complex binds to NF-κB to maintain a repressed state, which is terminated upon NCoR degradation. Ligand-dependent transrepression of PPARγ occurs when ligands induce PPARγ SUMOylation, which guides PPARγ to the HDAC3/NCoR complex, inhibits NCoR degradation, and causes NF-κB target gene repression (Pascual et al., 2005; Dekkers et al., 2012) (Figure 2B). As NF-κB is part of a prototypical pro-inflammatory signaling cascade, SUMOylation-dependent transrepression is the predominant means by which PPARγ promotes anti-inflammatory effects. Other mechanisms involved in PPARγ-induced NF-κB inhibition include ubiquitin-dependent degradation of NF-κB, exportation of NF-κB out of the nucleus, cofactor competition with NF-κB, and steric inhibition of NF-κB binding to DNA (Sauer, 2015) (Figure 2C). The sophisticated, multidimensional nature of PPARγ PTMs and functioning patterns confers this protein with the flexibility demanded of a master gatekeeper.

2.4 PPARγ target genes

The observation that PPARγ knockout is embryonically lethal indicates a vital and indispensible role for this protein (Semple et al., 2006). PPARγ response networks are complex and different genes are regulated in cell-specific manners. PPARγ regulates a variety of target genes relevant to adipogenesis (e.g. adipoq, lpl, nr1h3, ucp1), fatty acid metabolism (e.g. acadl, acadm, acox1), lipid transport (e.g. angptl4, apoe, olr1), cell proliferation (e.g. clu, eln, hspd1), insulin signaling (e.g. cpt1a, dgat1, pck1), and inflammation (e.g. nfkb, mmp9). The activation of PPARγ increases metabolic activities such as lipid metabolism and storage, adipogenesis, insulin sensitization, glucose homeostasis, and sodium and fluid retension (Ahmadian et al., 2013). Nevertheless, not all of these functions involve direct transcriptional regulation. The regulated genes can be grouped into three major categories (Chen et al., 2012): 1) fatty acid/glucose metabolism, 2) inflammation/oxidative stress/apoptosis, and 3) cancer. A large variety of genes are found in the area of overlap of these three categories and positive regulation is more common than negative regulation. Most of PPARγ signal transduction cascades in CNS pathologies are directly or indirectly associated with NF-κB, and this serves as a major mechanism whereby PPARγ regulates the stress response to injuries in the brain.

In addition to NF-κB, PPARγ also regulates the prototypical redox-sensing nuclear factor erythroid 2- related factor 2 (Nrf2) pathway, acting in synergy with this pathway to mute the destructive effects of oxidative stress (Figure 3) (Ikeda et al., 2000; Shih et al., 2005; Cho et al., 2010; Polvani et al., 2012; Zhao and Aronowski, 2014). Nrf2 is a master transcription factor that binds the antioxidant response element (ARE) and activates numerous genes critical for the maintenance of redox homeostasis, such as those related to the glutathione and superoxide dismutase antioxidant systems (Zhang et al., 2013). The interactions between PPARγ-Nrf2 signaling pathways can be classified into four categories. First, in addition to PPARγ-mediation modulation of the Nrf2/ARE axis, Nrf2 also regulates PPARγ, suggestive of a bidirectional loop (Cho et al., 2010). For example, Nrf2-mediated PPARγ induction protects mice against acute oxidant damage to the lung in vivo (Cho et al., 2010). Second, some antioxidant genes, such as catalase (Guan et al., 2000; Girnun et al., 2002), glutathione S-transferase (Rushmore and Pickett, 1990; Rushmore et al., 1991; Park et al., 2004a) and superoxide dismutase (SOD) (Yoo et al., 1999; Zelko et al., 2002; Jurkunas et al., 2010) contain both a PPRE and an ARE and are regulated by both PPARγ and Nrf2 to elicit anti-oxidative effects. Third, microglial/macrophagic CD36 is under the regulation of both PPARγ (Tontonoz et al., 1998; Yamanaka et al., 2012) and Nrf2 (Ishii et al., 2004; Wang et al., 2014; Zhao et al., 2015a). CD36 is important in microglial/macrophage phagocytosis and subsequent debris clearance, which facilitates resolution of neuroinflammation (Li et al., 2015). Fourth, PPARγ and Nrf2 synergistically elicit anti-inflammatory effects by inhibition of the NF-κB pathway (Wardyn et al., 2015; Zhao et al., 2015b).

Figure 3.

Figure 3

Interactions between PPARγ and Nrf2 pathways. (1) A bidirectional loop exists between PPARγ and Nrf2. PPARγ can upregulate Nrf2 expression and vice versa. (2) PPARγ and Nrf2 synergistically increase the expression of some antioxidants, including catalase, SOD, and GST, which have both PPRE and ARE elements in their genes. These proteins inhibit the generation of ROS, thereby mitigating neuroinflammation and improving debris clearance. (3) Microglial CD36 is regulated by both PPARγ and Nrf2. CD36 is critical for microglial phagocytosis and subsequent debris clearance, both of which facilitate the resolution of neuroinflammation. (4) PPARγ and Nrf2 synergistically inhibit NF-κB, the major proinflammatory pathway. Abbreviations: nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione S-transferase (GST), reactive oxygen species (ROS), superoxide dismutase (SOD).

PPREs have been found in target genes with potential roles in tissue preservation and repair and functional recovery after acute brain injury. Examples include tight junction protein zona occludens-3 (BBB integrity), synaptogyrin 4 (synaptic function), ubiquitin conjugating enzyme E2 S (protein quality control), G1/S-specific cyclin, p21-activated kinase 4, homeodomain-interacting protein kinase 4 (cell cycle), mitogen activated protein kinase organizer 1, transciption factor jun-D (cell survival), TOMM40, TIMM13, TIMM44, and electron transfer flavoprotein subunit beta (mitochondrial function) (Heinaniemi et al., 2007). Furthermore, a genome-wide identification of PPREs revealed functional clusters of PPAR-regulated genes related to chromatin remodeling, DNA damage response, cell differentiation, and cell growth/maintenance (Lemay and Hwang, 2006). Aside from the tissue reparative nature of these target genes, PPREs with well-established roles in lipid metabolism and inflammation might be critical for the repair of myelin and clearance of debris in the CNS. Indeed, a critical role for PPARγ in peripheral wound repair has been well established (Yessoufou and Wahli, 2010); PPARγ is activated by lipid mediators produced by necrotic tissue, and upregulates genes involved in anti-fibrotic, anti-oxidative, and anti-apoptotic effects (Michalik and Wahli, 2006). As discussed further below, the genomic studies identifying potential PPREs are consistent with the view that PPARγ lies at the nexus between perturbations of the internal milieu and a concerted effort to transcribe an abundance of genes that preserve homeostasis and repair tissue.

3. Evidence favoring a beneficial role of PPARγ in the injured CNS

CNS injuries encompass a variety of pathogenic mechanisms, including neurovasculopathy, trauma, neurodegeneration, tumor formation, and CNS autoimmunity, and may arise due to systemic diseases. In the present review, we will restrict our focus to those conditions with CNS abnormalities as the primary underlying pathology. This group of CNS injuries—ischemic stroke, hemorrhagic stroke, TBI, and SCI—share a number of pathophysiologic/repair processes in common. In the next section, we briefly describe alterations in PPARγ expression in CNS injuries and present evidence favoring its beneficial effects. We will then introduce the mechanisms underlying PPARγ-mediated protection against CNS injury, promotion of CNS repair, and regulation of comorbidities (Figure 4).

Figure 4.

Figure 4

Pathophysiology of CNS injuries and summary of PPARγ effects. The acute phase of CNS injuries (e.g. stroke, TBI, SCI, etc.) is characterized by neuroinflammation, neuronal death, demyelination, and BBB disruption. In the chronic recovery phase, repair processes are initiated, such as resolution of inflammation, clearance of debris and infiltrating cells, neurogenesis, remyelination, angiogenesis, and BBB repair. Comorbidities affect the risk of CNS injuries, and influence the course of the injury and self-reparatory activities. PPARγ inhibits the detrimental effects of injuries, promotes repair, and mitigates comorbidities. Abbreviations: blood brain barrier (BBB).

3.1 PPARγ expression and activity following CNS injuries

Ischemic stroke is caused by transient or permanent local reduction of cerebral blood flow, and is characterized by a number of cellular disturbances, including BBB dysfunction, vasogenic brain edema, and neuronal death (Yin et al., 2014; Krueger et al., 2015). In response to middle cerebral artery occlusion (MCAO), PPARγ mRNA levels are upregulated by 3.3-fold and 7.5-fold in the ipsilateral cerebral cortex of the rat at 6h and 24h post-MCAO, respectively (Ou et al., 2006). In situ hybridization studies further reveal that PPARγ mRNA expression is restricted to neurons within the ischemic territory (Ou et al., 2006). Similarly, Victor et al. observed PPARγ upregulation at both the mRNA and protein levels in ischemic neurons following MCAO in rats (Victor et al., 2006). Elevated expression of PPARγ protein was detected by immunohistochemistry as early as 4h after stroke, with a peak at 24h, and lasted for at least 14d after stroke (Victor et al., 2006). Despite increased PPARγ expression in neurons after ischemic insults, there is decreased PPARγ DNA binding activity in the ischemic hemisphere. In the ischemic brain, PPARγ-PPRE binding activity 8 h after the insult is only ~44% of the values in the contralesional brain, as evaluated by electrophoretic mobility shift assays and densitometry (Victor et al., 2006). The reason for the reduction in the PPRE binding activity of PPARγ after ischemic injury is still unknown. It is possible, however, that PPARγ protein levels may be elevated in the ischemic brain to compensate for the decrease in PPARγ DNA binding activity.

Hemorrhagic stroke is elicited by the rupture of a weakened blood vessel or a brain aneurysm followed by bleeding into surrounding brain regions. There are two subtypes of hemorrhagic stroke—intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH) (Qureshi et al., 2001; Tholance et al., 2015). Xu et al. injected autologous whole blood into the right striatum of male Sprague-Dawley rats to induce an ICH model (Xu et al., 2016). Significantly elevated expression of PPARγ and 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) was found in the ipsilateral brain in ICH rats compaired to sham-treated controls (Xu et al., 2016). Furthermore, PPAR-γ protein levels and PPARγ-targeted gene expression were significantly increased in rats treated with the PPARγ agonist, exogenous 15(S)-HETE, in comparison to vehicle-treated ICH rats (Xu et al., 2016). Intrastriatal injections of 15d-PGJ2 significantly increased PPARγ-DNA binding activity and the expression of catalase in the perihemorrhagic area 3h after ICH (Zhao et al., 2006). However, in an experimental SAH model, PPARγ mRNA and protein levels were significantly reduced in the cortex compared to sham controls (Chang et al., 2015a).

Following TBI, cerebral inflammation is one of the major triggers of progressive neuronal cell death (Morganti-Kossmann et al., 2001). In a TBI model induced by controlled cortical impact (CCI), PPARγ mRNA expression was not altered either following TBI or treatment with the PPARγ agonists PGZ and RGZ (Thal et al., 2008; Thal et al., 2011). However, inflammatory genes such as inducible nitric oxide synthase (iNOS), TNF-α, interleukin (IL)-1β and IL-6 were upregulated following TBI, and this response was inhibited by PGZ treatment (Thal et al., 2011). In contrast, Yi et al. reported a two-fold increase in PPARγ mRNA expression 24h after experimental TBI and this was increased four-fold by RGZ treatment (Yi et al., 2008). The methodological differences in experimental trauma models might explain these discrepancies.

It is well accepted that apoptosis, inflammation, and excitotoxicity contribute to secondary neuronal damage after SCI. One day following experimental SCI, the expression of PPARγ is significantly increased in the spinal cortex surrounding the lesion area (Zhang et al., 2010). In contrast, another study demonstrated that PPARγ protein levels were significantly reduced in spinal cord homogenates one day after SCI (Paterniti et al., 2013).

3.2 Protective effects of PPARγ in CNS injuries

A large body of seminal work in healthy control, diabetic, or obese animals revealed that PPARγ agonists decrease myocardial infarct size and increase glucose uptake and insulin sensitivity (Yue Tl et al., 2001; Khandoudi et al., 2002; Shiomi et al., 2002; Sidell et al., 2002; Wayman et al., 2002; Lee and Chou, 2003; Liu et al., 2004). Consistent with these studies, mounting evidence also demonstrates that PPARγ activation is beneficial to the injured brain (Zhao and Aronowski, 2014). For example, Zhao and colleagues demonstrated that neuron-specific PPARγ knockout mice experience greater brain damage and oxidative stress after MCAO (Zhao et al., 2009b). Furthermore, systemic or intracerebroventricular administration of PPARγ agonists, either prior to and/or after stroke, elicit potent neuroprotective effects in various animal models of ischemic stroke, as shown by reductions in infarct size and superior neurological function (Sundararajan and Landreth, 2004; Luo et al., 2006; Wang et al., 2009; Kaundal and Sharma, 2010). Even delayed intraperitoneal administration of the PPARγ agonist RGZ—beginning 24 h after cerebral ischemia—is able to confer protection in a middle cerebral artery embolization stroke model in rats (Allahtavakoli et al., 2009). These findings may be important because they suggest a wide therapeutic window for PPARγ agonists. However, the dose must be chosen with care, as PPARγ agonists may promote cell survival at low concentrations but increase cell loss at high concentrations (Wang et al., 2002b; Lin et al., 2006; Wu et al., 2009).

Aside from ischemic stroke, PPARγ agonists also exhibit neuroprotective effects in hemorrhagic stroke models (Zhao et al., 2006; Zhao et al., 2007; Zhao et al., 2009a; Wu et al., 2015). For example, in a rat model of striatal ICH, intrastriatal 15d-PGJ2 treatment increased PPARγ DNA binding activity and subsequent catalase expression, resulting in significant decreases in neurologic deficits compared to rats receiving saline (Zhao et al., 2006). Similarly, intraperitoneal administration of RGZ at 5 min, 6 h, and 24 h after TBI increased PPARγ mRNA levels in cortical tissue surrounding the contusion area within 24 h after brain injury and significantly reduced the cortical lesion volume compared to vehicle-treated mice (Yi et al., 2008). Similarly, the survival of hippocampal CA3 neurons was increased after intraperitoneal injection of 6 mg/kg RGZ in a controlled cortical impact model of TBI (Liu et al., 2016). Nevertheless, a number of studies have shown contradictory results, particularly in TBI models. It has therefore been argued that the neuroprotective effects of PGZ in TBI—as shown by reduced contusion volume, decreased cerebral inflammation, and reduced microglial activation—are the result of PPARγ-independent mechanisms (Thal et al., 2011; Liu et al., 2016). Other possible reasons for these discrepancies include: 1) differing rodent species and strains; 2) varying degrees of injury severity, reflecting differences in the diameter of the impactor tip, impact velocity, impact duration, and displacement; 3) differences in lesion location within the brain; 4) different administration methods and timing of PPARγ agonists; and 5) different times of evaluation. In short, the interpretation of this body of work awaits further confirmation and greater reproducibility across studies.

Application of PPARγ agonists is protective in various animal models of SCI. In a study by McTigue and colleagues, rats received a moderate mid-thoracic contusion and were intraperitoneally injected with either low or high doses of PGZ or vehicle at 15 min after SCI and then every 12h for 7 days (McTigue et al., 2007). In this study, PGZ significantly protected white matter, gray matter, and motor neurons from SCI. In a compression model of SCI, vascular clips were applied to the mouse dura with a force of 24g via four-level T5-T8 laminectomy (Genovese et al., 2008). In this model, intraperitoneal injections of the PPARγ agonist 15d-PGJ2 ameliorated spinal cord inflammation, tissue injury, neutrophil infiltration, NF-κB activation, and cell apoptosis after SCI. In addition, the PPARγ agonist PGZ was found to ameliorate sensory dysfunction after SCI (Iwai et al., 2008) and reduce neuropathic pain after SCI and peripheral nerve injury (Jia et al., 2013; Griggs et al., 2015). Although there remain some debates, the abovementioned findings suggest that PPARγ agonists elicit structural and functional protection of the brain in diverse injury models.

4. PPARγ protects against CNS injury

A series of pathological processes are initiated during the acute phase following CNS injuries. Following the acute period, a wave of secondary injury expansion also emerges. Necrosis of neural tissue triggers a powerful inflammatory cascade, resulting in further neuronal apoptosis and excitotoxicity, oxidative stress, and other feed-forward mechanisms. Neuroinflammation contributes to the amplification and spread of local injury, predisposing surrounding cells to secondary injuries (Kawabori and Yenari, 2015). Vulnerable brain vessels and the BBB release pro-inflammatory factors and facilitate immune cell infiltration into the injured brain (de Wit et al., 2017). In this section, we will summarize pathophysiological processes after CNS injury and the beneficial effects of PPARγ in these processes.

4.1 PPARγ protects against neuroinflammation following CNS injuries

The immune system serves as the sentinel and guardian of the organism. For many decades, the CNS was falsely believed to be immunoprivileged. The CNS is now viewed as immune competent, actively engaging in interactions with the peripheral immune system (Carson et al., 2006). Furthermore, a lymphatic vascular system has been discovered in the brain (Molina-Holgado and Molina-Holgado, 2010; Aspelund et al., 2015) and neural inflammation is thought to drive disease processes into a secondary phase of injury expansion.

4.1.1 An introduction on neuroinflammation following CNS injuries

Inflammatory responses following CNS injury are initiated by damage-associated molecular patterns (DAMPs) that are released from injured tissue (Sharma and Naidu, 2016). High mobility group box 1 is rapidly released by cells undergoing necrotic or pyroptotic cell death (Sharma and Naidu, 2016). High mobility group box 1 is detected in the serum half an hour after 90 min stroke in rodents, and is observed within the first hour after symptom onset in ischemic stroke patients (Liesz et al., 2015). In addition, heat shock protein (HSP) 72 can be detected in the serum of CNS trauma patients (Pittet et al., 2002). Release of S-100 protein from the injured CNS occurs within the first 24h in patients with TBI, ischemic stroke, and transient ischemic attacks (Elting et al., 2000). Furthermore, the early release of nuclear DNA, the IL-1 family, histones, and other intrinsic cell components from the injured CNS serves to ignite the inflammatory response after acute CNS injuries (Sharma and Naidu, 2016).

Both local and systemic inflammatory cells participate in acute CNS injury. Microglia are the resident immune cells of the brain and are among the first cells to respond to CNS injury. Injury signals are transported through the CNS lymphatic vessels and the blood vessels, possibly delivered by the drainage of damage-associated molecular patterns directly to peripheral immune organs (Louveau et al., 2015). Injury signals might also be carried by dendritic cells (DCs) (Clarkson et al., 2012). Macrophages take up residence in the CNS vessel bed and are also poised to sense and transmit information about disruptions in homeostasis (Goverman, 2009; Kivisakk et al., 2009). Activation of the peripheral immune response results in a surge of infiltrating immune cells that home in on the CNS lesion zone (Pennypacker and Offner, 2015).

In ischemic stroke, the pro-inflammatory profiles of microglia may be inactivated at early stages after ischemic stroke. However, as the immune reactions progress, pro-inflammatory microglia participate in the promotion of neural inflammation (Hu et al., 2012; Patel et al., 2013; Wang et al., 2013; Jin et al., 2017). In a transient MCAO model, brain infiltration of macrophages appeared as early as 12h after ischemia and peaked after 24h, followed by a gradual decrease by day 7 (Gelderblom et al., 2009). Similarly, infiltrating neutrophils were detected at 12 h, peaking on day 2-3 and gradually disappearing by day 7 (Gelderblom et al., 2009). Although a deleterious role has been reported for natural killer cells in ischemic stroke (Gan et al., 2014), the infiltration of this cell population is not always evident. However, T and B lymphocytes from the adaptive immune systems appear in the ischemic lesion site at 3-7 days after stroke (Gelderblom et al., 2009), even though they represent only a small portion of infiltrated immune cells. In animal models of hemorrhagic stroke, microglial activation is observed 1-4 h after hemorrhage, peaking at 3-7 days, and returning to baseline by 21-28 days (Hickenbottom et al., 1999; Gong et al., 2000; Xue and Del Bigio, 2000; Wang et al., 2003a; Wang and Tsirka, 2005). Neutrophil infiltration occurs as early as 4h after the formation of the hematoma, peaking at 2-3 days, and subsiding by 7 days.

In traumatic CNS injuries such as TBI and SCI, the BBB is directly disrupted by the primary damage, followed by the entry of blood components into the CNS (McKee and Lukens, 2016). As in peripheral injured tissue, neutrophils are the first to be mobilized to the brain after TBI. In a rat model of CCI, neutrophil infiltration was detected within hours post-trauma, and this further exacerbated the loss of BBB integrity. Microglia and macrophages are increased between 12 and 72 h in damaged cortical regions after TBI (Clark et al., 1994; Soares et al., 1995). A large number of chemokines are released into the injured CNS, inducing the migration of activated neutrophils, lymphocytes, monocytes, and macrophages (Ghirnikar et al., 1998).

Together with the resident microglia in the CNS, infiltrated peripheral immune cells promote ongoing neural inflammation with direct cytotoxicity or secretion of inflammatory factors. Microglia produce proinflammatory cytokines such as IL-1, IL-6, IL-12, IL-23, nitric oxide (NO), and TNFα, and contribute to local oxidative stress by inducing iNOS and generating reactive oxygen species (ROS) (Hu et al., 2012; Wang et al., 2013). CD8+ cell cytotoxic T lymphocytes (CTLs) and natural killer cells produce cytolytic granzyme-b (Chaitanya et al., 2011) and perforin (Mracsko et al., 2014) to induce direct neuronal death after ischemic stroke. Immune cell-derived cytokines, including IFN-γ, TNF-α, various interleukins, macrophage inflammatory protein 1α and 1β, and monocyte chemotactic protein (MCP)-1β not only convey immune signals but also induce apoptosis of cells in the stroke penumbra (Jin et al., 2010b; Gan et al., 2014). In addition, chemokines are produced in the inflamed zone of injury, both by infiltrating leukocytes and the injured brain tissue itself, in order to attract even more peripheral immune cells in a self-amplifying cascade (Losy et al., 2005; Jin et al., 2010b; Wang et al., 2012b; Wolinski and Glabinski, 2013). Matrix metalloproteinases (MMPs) are released mainly from neutrophils and cause damage to the BBB, facilitating the entry of additional peripheral leukocytes. In addition, activated endothelial cells in microvessels express adhesion molecules that promote the rolling and firm adhesion of leukocytes and platelets (Yilmaz and Granger, 2008). It is worth noting that the deleterious effects of the aforementioned inflammatory factors are relatively non-specific, resulting in broad injury to cells in the lesion zone and surrounding tissues, including neurons, white matter-forming oligodendrocytes, and all components of the BBB.

4.1.2 PPARγ protects against post-injury neuroinflammation

Widespread expression of PPARγ has been detected in various immune cells, including CNS glial cells (Bernardo et al., 2000) and peripheral leukocytes (Ricote et al., 1998; Clark et al., 2000; Faveeuw et al., 2000; Padilla et al., 2000; Zhang et al., 2004a). A strong suppressor of the pro-inflammatory transcriptional factor NF-κB, PPARγ fine-tunes neural inflammation by limiting the activation of immune cells, reducing the generation of pro-inflammatory cytokines, chemokines, and adhesion molecules, and mitigating oxidative stress (Kapadia et al., 2008).

Treatment with PGZ reduces microglial proliferation in stroke-prone renovascular hypertensive rats (Lan et al., 2015). In addition, RGZ decreases parenchymal accumulation of neutrophils in a mouse MCAO model (Luo et al., 2006) and 15d-PGJ2 inhibits neutrophil infiltration following ICH injury (Zhao et al., 2006). In rodent TBI models, RGZ reduces activated microglia/macrophages, and this can be reversed by GW9662 (Yi et al., 2008). In vitro experiments reveal that RGZ impedes the maturation of human monocyte-derived DCs (Gosset et al., 2001). Although it is not known if lymphocyte activation is affected by PPARγ, the tempering of DC activation suggests this to be the case.

A large body of evidence indicates that activation of PPARγ attenuates neural inflammation by reducing inflammatory mediators in ischemic stroke (Culman et al., 2007), ICH (Zhao et al., 2007; Aronowski and Zhao, 2011), TBI (Qi et al., 2010), and SCI (Zhang et al., 2010; Li et al., 2013). PPARγ stimulation inhibits the expression of a wide array of pro-inflammatory mediators in microglia and macrophages, including TNF-α, IL-1β, IL-6, iNOS, inducible cyclooxygenase (COX) 2, and the IL-12 family (Bernardo et al., 2000; Luna-Medina et al., 2005; Storer et al., 2005). Appel et al. reported that 15d-PGJ2 or TGZ treatment of magnet-sorted human DCs (stimulated with toll-like receptor ligands) significantly down-regulated the production of cytokines (IL-12, IL-6, TNF-α, etc.) and chemokines (CCL2, CCL3, CCL5) that are involved in T cell recruitment and activation (Appel et al., 2005). Furthermore, PPARγ agonists have been shown to reduce the expression of IFNγ (da Rocha Junior et al., 2013), IL-17, and IL-23 in T cells (Klotz et al., 2009). In human endothelial cells, adenovirus-mediated expression of a constitutively active, mutant version of PPARγ suppressed the expression of vascular adhesion molecules intracellular adhesion molecule-1, vascular cell adhesion molecule-1 and E-selectin (Wang et al., 2002a).

Severe neural inflammation following acute CNS injury causes apoptosis of neurons (Aktas et al., 2007), degeneration of white matter (Rosenberg, 2009), and breakdown of the BBB (Hawkins et al., 1991). PPARγ activation ameliorates these consequences of neural inflammation and oxidative stress in various acute CNS diseases (Culman et al., 2007; Qi et al., 2010; Zhang et al., 2010; Aronowski and Zhao, 2011). However, PPARγ may also be essential for microglial survival (Liu et al., 2010; Kane et al., 2011; Pan et al., 2013). Furthermore, a PPARγ agonist has been shown to restore compromised pro-inflammatory responses in diabetic mice soon after stroke, whereas inflammatory responses were suppressed at later time points in both control and diabetic mice (Kumari et al., 2010). These data demonstrate a sophisticated, bidirectional regulation of microglia by PPARγ.

4.2 Neuronal death

4.2.1 Mechanisms of neuronal death following CNS injury

CNS injury-associated neuronal death can be classified as primary or secondary. Primary neuronal death arises from energy failure in ischemic stroke, mechanical damage associated with mass effects in intracerebral hemorrhage (Qureshi et al., 2009), and mechanical damage from blunt force traumas in TBI and SCI (McTigue, 2008). The loss of neurons typically occurs immediately following injury and continues for hours to days. An important feature of primary neuronal death is that it is generally believed to be non-regulatable and non-intervenable. On the contrary, secondary neuronal death may be regulated with interventions and is therefore the focus of the following discussion.

Glutamate receptors belong to two major classes—G-protein coupled metabotropic receptors and ligand-gated ionotropic receptors, such as the N-methyl-D-aspartic acid (NMDA) receptor, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor, and kainate receptor. Excitotoxicity is induced by excessive activation of glutamate receptors by extracellular glutamate and is a major contributor to neuronal death following CNS injury (Puyal et al., 2013). Energy failure after ischemia leads to decreased ATP production, thereby disrupting Na+/K+-ATPase, Ca2+/H+-ATPase and Na+/Ca2+-transporters and leading to depolarization of the cellular membrane. Diffusely depolarized presynaptic neurons release excessive glutamate into the synaptic cleft (Terasaki et al., 2014; Hamming et al., 2016). Another mechanism of excessive glutamate release is via connexin hemichannels in activated astrocytes and microglia (Takeuchi et al., 2006; Ouyang et al., 2014). Ultimately, over-activation of the NMDA receptor leads to the sustained excitement of neurons and an excessive influx of calcium, which causes loss of neuronal viability (Randall and Thayer, 1992; Tymianski et al., 1993; Owens et al., 1997; Lai et al., 2014).

Aside from excitotoxicity-induced neuronal cell death, oxidative stress may also elicit cell loss in CNS injury conditions. The predominant ROS are the superoxide anion (O2), hydrogen peroxide (H2O2), hydroxyl radical (OH), peroxyl (RO2), peroxynitrite anion (ONOO), and nitrogen dioxide (NO2) (Dhawan, 2014; Yang et al., 2016). Excessive intracellular Ca2+ accumulation results in overactivation of calcium-dependent enzymes such as nicotinamide adenine dinucleotide phosphate oxidase (NOX) and xanthine oxidase, both of which are capable of producing ROS. Disruptions in the electron transport chain also elicit the leakage of ROS from mitochondria (Piantadosi and Zhang, 1996; Galeffi et al., 2016). Other mechanisms of ROS production include lipid peroxidation and increased mitochondrial ROS production stimulated by the release of Ca2+, Na+, and adenosine diphosphate (ADP) from damaged cells (Schulz et al., 2000; Viola and Hool, 2013). Furthermore, blood components, particularly hemoglobin, can undergo auto-oxidation upon erythrocyte lysis, which may serve as a major source of ROS in ICH, SAH, TBI, and SCI (Simoni et al., 1990; Asano, 1999). ROS are typically released within 1h post-injury in diverse models (Gaetani et al., 1990; Marklund et al., 2001). The cellular source of this oxidative stress might be neurons, other parenchymal cells, and vascular cells (Chen et al., 2014). ROS can directly damage neuronal components by promoting lipid peroxidation, protein breakdown, and DNA damage, and also act as executioners of cell death by activating pro-apoptotic pathways (Zhang et al., 2004b). Finally, ROS can promote neuroinflammation, BBB disruption, and the release of vasospastic mediators that indirectly harm neurons by induction of leukocyte infiltration and enhancement of inflammation, thereby exacerbating ischemic injury (Clark et al., 1993).

4.2.2 PPARγ protects against neuronal death following CNS injury

PPARγ has been shown to decrease excitotoxicity in various pathologic settings. For example, 15d-PGJ2 protects neurons against NMDA-induced toxicity both in vivo and in vitro (Zhao et al., 2006). Similarly, the PPARγ agonist RGZ promotes neuronal survival and neurite outgrowh in vitro following exposure to glutamate (Zuhayra et al., 2011). In a rodent ischemic stroke model, the PPARγ antagonist T0070907 abolished neuroprotection mediated by ischemic preconditioning, and increased glutamate release and reduced glutamate uptake in neuron-astrocyte co-cultures (Romera et al., 2007). Similar effects have also been reported in other disease models. TGZ diminishes glutamate-induced excitotoxicity to the same degree as NMDA receptor antagonists in retinal ganglion cells (Aoun et al., 2003). Furthermore, RGZ decreases glutamate release in glioblastoma multiforme, and this can be reversed by the PPARγ antagonist GW9662 (Ching et al., 2015). The underlying mechanism by which PPARγ reduces excitotoxicity may involve upregulation of the glutamate transporter (GLT)-1 (Romera et al., 2007; Ching et al., 2015). GLT-1 is found in astrocytes and is responsible for up to ~90% of glutamate uptake (Robinson, 1998; Schousboe and Waagepetersen, 2005; Arbo et al., 2016). On the other hand, Yao et al observed no changes in GLT-1 expression in response to RGZ injection in a model of TBI (Yao et al., 2015). In addition, no evidence supporting the effects of PPARγ on excitotoxicity has been reported in SCI and hemorrhagic stroke models. Nevertheless, increased astrocytic glutamate uptake following PPARγ activation would be expected to reduce overall tissue damage (Romera et al., 2007; Ching et al., 2015), as excitotoxicity is a major cause of death not only for neurons, but also for oligodendrocytes and other types of CNS cells (Huria et al., 2015).

The anti-oxidative effects of PPARγ are better established than its effects on excitotoxicity. First, PPARγ can directly inhibit ROS generation, partly by inhibiting ROS-generating enzymes. RGZ has been shown to promote neuronal survival and neurite outgrowh in vitro following exposure to the Parkinson’s oxidative toxicant 6-hydroxydopamine (Zuhayra et al., 2011). Systemic administration of RGZ or PGZ after ischemia/reperfusion reduces ROS and the expression of COX-2, an important enzyme involved in ROS generation, and attenuates neuronal loss (Collino et al., 2006). Similarly, 15d-PGJ2 inhibits iNOS expression and subsequent apoptosis in cerebellar granule cells (Heneka et al., 1999). NOX activation is another major driving force in ROS generation, and its relationship with PPARγ has been examined in primary cortical neurons subjected to oxygen-glucose deprivation (OGD) (Wu et al., 2016). NOX expression was enhanced by both GW9662 treatment and PPARγ-siRNA transfection, and was inhibited by PPARγ transfection or exposure to 15d-PGJ2. Second, PPARγ prevents oxidative stress by induction of anti-oxidant molecules. For example, both RGZ and PGZ can restore glutathione, the most ubiquitous biological antioxidant, in a mouse MCAO model (Collino et al., 2006). Furthermore, in a rodent TBI model, RGZ induces the expression of the antioxidant enzymes catalase and SOD-1 (also known as Cu/Zn SOD), as well as heme oxygenase-1 and the chaperones heat shock protein (HSP) 27 and HSP70, and these responses are abolished by GW9662 administration (Yi et al., 2008). As mentioned above, PPARγ also interacts with the Nrf2/ARE axis to mute the toxic sequelae of oxidative stress (Ikeda et al., 2000; Shih et al., 2005; Cho et al., 2010; Polvani et al., 2012; Zhao and Aronowski, 2014). Collectively, these findings reveal the molecular mechanisms underlying powerful anti-oxidative actions of PPARγ. As oxidative stress contributes to almost all CNS pathologies and impairs tissue repair, the upregulation of multiple antioxidant pathways by PPARγ is expected to accelerate the recovery process.

Another mechanism underlying PPARγ-afforded neuroprotection may be through the energetic and neurotrophic support provided by glial cells. Production of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) in CNS trauma models is enhanced after administration of PPARγ agonists, although the cellular source of the neurotrophins has not been identified (Meng et al., 2011; Mandrekar-Colucci et al., 2013). Astrocytes have long been known to provide nutritional and metabolic support to neurons (Wang et al., 2016), and activated astrocytes produce BDNF following injury (Zhao et al., 2004). Consistent with its established role in bioenergetics, PPARγ modulates glucose metabolism in astrocytes. For example, PPARγ agonists increase glucose uptake and glycolysis in primary astrocytes (Dello Russo et al., 2003; Izawa et al., 2009). The subsequent increase in cerebral glucose utilization and metabolism might attenuate neurodegeneration (Garcia-Bueno et al., 2007). These effects might be especially important in diseases related to energy deprivation—such as ischemic stroke—and may broaden the temporal window during which drugs might be administered, in addition to increasing cell survival. Microglia also secrete trophic factors (e.g. insulin-like growth factor (IGF)-1 and BDNF) to support neuronal survival (Choi et al., 2008; Trang et al., 2011; Wang et al., 2017). However, it is not known if PPARγ activation increases trophic factor expression in microglia.

4.3 Demyelination

4.3.1 Mechanisms of demyelination following CNS injury

White matter injury is characterized by demyelination and axonal degeneration. Demyelination is defined as loss of the axonal myelin sheath that is formed by the processes of oligodendrocytes. Myelination is essential for efficient signal transduction across different brain regions, and serves to protect the ensheathed axon by providing metabolic support and acting as a physical barrier against biochemical insults (Irvine and Blakemore, 2008; Funfschilling et al., 2012; Lee et al., 2012). Axonal degeneration can ensue as a consequence of demyelination, and these secondary effects are largely responsible for the debilitating effects of white matter injury. Thus, the consequences of demyelination include sensorimotor dysfunction and profound neurobehavioral and cognitive impairments (Desmond, 2002).

Oligodendrocytes in white matter are highly vulnerable to ischemic injury for a number of reasons. First, white matter has much lower blood flow and lack of collateral circulation in comparison with gray matter (Rosenberg, 2016). Second, cells of the oligodendrocyte lineage, such as oligodendrocyte precursor cells (OPCs), are particularly vulnerable to ischemia-induced oxidative stress and inflammation (Husain and Juurlink, 1995). In the rat MCAO model, 30 min of vascular occlusion leads to conspicuous swelling of oligodendrocytes and 3 h of occlusion results in lethal injury in large numbers of oligodendrocytes (Pantoni et al., 1996). Demyelination persists for at least 4 weeks after 60 min of transient MCAO in mice, as evidenced by decreased expression of myelin basic protein (MBP) in white matter (Jiang et al., 2016). Consistent with these in vivo observations, in vitro studies show that 30 min of OGD results in the death of 90% of oligodendrocytes within 9h after reperfusion (Tekkok and Goldberg, 2001). The pattern of demyelination in other CNS injuries, such as intracerebral hemorrhage, SCI, and TBI is similar to that observed in ischemic stroke. In other words, demyelination commences within hours to days after injuries and is sustained for a prolonged period, often up to months or even years (Wasserman and Schlichter, 2008).

The molecular mechanisms underlying CNS injury-induced demyelination and oligodendrocyte death can be summarized as follows. First, oligodendrocytes express all three types of glutamate receptors—AMPA, kainate, and NMDA (Salter and Fern, 2005; Butt, 2006), and are therefore subject to excitotoxicity. As in neurons, excess extracellular glutamate leads to over-activation of glutamate receptors on oligodendrocytes and elicits the accumulation of cytosolic calcium, which triggers oligodendrocyte toxicity (Benarroch, 2009). Second, oxidative stress is another major contributor to demyelination. Oligodendrocytes have high energy demands, abundant iron stores, and low levels of molecular antioxidants, rendering them particularly sensitive to increased ROS (Juurlink, 1997; Oyinbo, 2011; Shereen et al., 2011; Yang et al., 2017). Third, inflammatory reactions also contribute to demyelination after ischemia. Activation of resident microglia and astrocytes, as well as the infiltration of peripheral immunocytes lead to excess production of proinflammatory cytokines, key mediators of pathology in demyelination disorders. For example, high levels of IFN-γ lead to oligodendrocyte apoptosis and demyelination (Lin et al., 2005). Furthermore, TNF-α inhibits the proliferation and differentiation of OPCs, and induces oligodendrocyte apoptosis (Hovelmeyer et al., 2005; Pang et al., 2005; Shi et al., 2015).

Interactions between oligodendrocyte lineage cells and other white matter components such as microglia, astrocytes, and axonal fibers also play a critical role in injury-induced demyelination. Activated microglia can exhibit pro-inflammatory and anti-inflammatory properties, respectively (Hu et al., 2015) and may influence oligodendrocyte survival (Wang et al., 2013). Similarly, activated astrocytes also exhibit dual roles. Glutamate is taken up by astrocytes and converted into lactate, which then serves as an energy substrate for axons and oligodendrocytes following intracellular entry through monocarboxylate transporters and connexins. Astrocytes thereby transiently provide energy for oligodendrocytes and axons under stressful conditions (Matute et al., 2007). However, activated astrocytes may exacerbate oligodendrocyte cell death by increasing intracellular uptake of zinc and stimulation of Ca2+-permeable AMPA receptors (Johnstone et al., 2013).

Traumatic axonal injury (TAI) is localized to white matter and is initiated by mechanical force, such as impact to the head and rapid acceleration-deceleration of the head or the spinal cord (Armstrong et al., 2016b). The proliferation, migration, survival, and differentiation of oligodendrocytes depend upon axon-derived signals (Shi et al., 2015), such that loss or degeneration of axons can directly cause myelin pathologies. TAI causes axonal degeneration, which is accompanied by mitochondrial swelling, disruption of vesicular transport, cytoskeleton breakdown, and the formation of end bulbs at the sites of axotomy or disconnection. An important feature of TAI is that the injury is dispersed among the intact axons within white matter tracts; this expands the lesion and increases disease severity (Gennarelli, 1993). Mild TBI with low TAI density does not impair oligodendrocyte survival, as a single oligodendrocyte can maintain myelin sheaths around a cohort of axons, and damaged axons are a minor proportion of the total number of axons present. Nevertheless, damage to axons can dysregulate myelin maintenance signals to the oligodendrocyte, resulting in aberrant myelin synthesis and subsequent formation of redundant myelin, as evident in double-layered myelin sheaths around damaged axons (Rosenbluth, 1966; Armstrong et al., 2016a). Mild TBI may be accompanied by some degree of demyelination, as there also co-exist other pathogenic factors related to demyelination, including glutamate excitotoxicity, excessive ROS, neuroinflammation, and microglial activation (Clarner et al., 2012; Sullivan et al., 2013; Mierzwa et al., 2015). Consistent with these observations, in a closed-skull impact mouse model mimicking mild TBI, demyelination of intact axons is observed as early as 3 days, and lasts up to 6 weeks post-injury, in the absence of frank oligodendrocyte death (Mierzwa et al., 2015). In that model, chronic inflammation contributes to the long-lasting demyelination. Moderate or severe TBI with TAI is often accompanied by microhemorrhages or severe neuroinflammation (Armstrong et al., 2016a), resulting in focal lesions, and a higher proportion of axons in the lesion site are damaged, leaving the myelinating oligodendrocytes without any viable axons to wrap (Armstrong et al., 2016b). This lack of nearby axons causes myelin degradation and oligodendrocyte death. It is worth noting that TBI and SCI also cause mechanical damage in gray matter, leading directly to neuronal death (Bayly et al., 2005). Thus, axons can become disconnected from damaged neurons and undergo orthograde degeneration, ultimately leading to myelin degradation and oligodendrocyte death, as outlined above. For these and other reasons, death of oligodendrocytes in white matter is evident within 12 h in a rat model of moderate fluid percussion injury, and this increases progressively through the following week and is maintained for 2 months post-injury (Conti et al., 1998), perhaps mediated by the caspase-3 apoptotic pathway (Flygt et al., 2013).

4.3.2 PPARγ protects against demyelination following CNS injury

Most studies on PPARγ in CNS injury focus on gray matter. In a recent study, however, Han et al. investigated the effect of PGZ on white matter integrity in a 60 min transient MCAO mouse model, and reported less loss of white matter integrity at 21d post injury, partly due to increased microglial M2 polarization (Han et al., 2015). There are also protective effects of PPARγ on autoimmunity-induced demyelination, consistent with its beneficial role in stroke, TBI, and SCI. In rodent experimental autoimmune encephalomyelitis (EAE) models, TGZ decreases pathology by attenuating pro-inflammatory cytokine production (Niino et al., 2001). Similarly, PGZ reduces injury symptoms in EAE models, and this is accompanied with decreased lymphocyte infiltration, demyelination, and chemokine and cytokine expression, and increased expression of the NF-κB inhibitor, IκB, in the brain (Feinstein et al., 2002).

In vitro studies have demonstrated potential mechanisms underlying the protective effects of PPARγ against demyelination. For example, endogenous prostaglandin derivatives are natural PPARγ activators and inhibit production of nitrite and pro-inflammatory cytokines, including IL-6, IL-1β, TNF-α, and MCP-1 from LPS-stimulated microglia and astrocytes (Storer et al., 2005). In a study exploring T-cell mediated demyelination in three-dimensional aggregating rat brain cell cultures, Duvanel et al. demonstrated that PGZ rescued anti-myelin oligodendrocyte glycoprotein autoantibody-induced demyelination by reducing heat shock responses and downregulating TNF-α (Duvanel et al., 2003). Apart from its anti-inflammatory properties, PPARγ also mitigates oxidative stress in oligodendrocytes. Treatment of rat oligodendrocytes with PGZ or 15d-PGJ2 enhances their antioxidant defenses by increasing levels of catalase and Cu/Zn SOD, while maintaining the equilibrium of the glutathione redox system (Bernardo et al., 2009). Although there is little direct evidence regarding the influence of PPARγ on excitotoxicity-induced demyelination, PPARγ is known to decrease excitotoxicity in general, as discussed in section 4.2.2. However, whether PPARγ can help relieve demyelination caused by TAI-induced oligodendrocyte death is not yet known.

Collectively, the available evidence supports the therapeutic potential of PPARγ in CNS injury- induced demyelination, although evidence regarding direct effects of PPARγ on demyelination is still limited, especially in the context of acute CNS injuries. Furthermore, the underlying mechanisms are poorly understood and warrant further studies.

4.4 BBB disruption

4.4.1 Mechanisms of blood-brain barrier disruption in CNS injury

As a physical, transport, and metabolic barrier, the blood-brain barrier (BBB) controls the exchange of various molecules and nutrients between the blood and brain compartments, while keeping bacteria, viruses, and other xenobiotics outside the brain (Weiss et al., 2009). The major structural bases of the BBB consist of brain capillary endothelial cells, the tight junctions that connect them, astrocyte end feet, and pericytes (Weiss et al., 2009; Thal and Neuhaus, 2014; Attwell et al., 2016). Rapid alterations of endothelial permeability occur in many neurological conditions, including ischemic stroke and TBI (Salehi et al., 2017). The effects of CNS injuries may occur either directly on the endothelium or indirectly through other cells types in the neurovascular unit that affect the endothelium (Thal and Neuhaus, 2014). Reactive oxidants, inflammatory cytokines, and MMPs contribute to BBB permeability and cause the degradation of endothelial junctions, loss of vascular integrity, and BBB dysfunction after CNS injuries. Within 30-60 min following ischemic stroke, aberrant actin polymerization, stress fiber formation, and destruction of junctional proteins in brain endothelial cells result in an early breach of the BBB in response to activation of Rho-associated protein kinase/myosin light chain signaling and loss of inhibition by actin depolymerizing factor. This is followed by the infiltration of MMP-secreting neutrophils and macrophages into the brain, setting in motion a cascade of events that culminates in tissue damage and loss of neurological function (Shi et al., 2016a; Shi et al., 2016b). Increased BBB permeability may also facilitate edema formation and hemorrhagic transformation, thereby aggravating neurological outcomes (Dirnagl et al., 1999). All of these changes lead to primary brain damage and post-ischemic secondary injury (Hacke et al., 1996; Yin et al., 2010).

4.4.2 PPARγ protects against blood-brain barrier disruption following CNS injury

The BBB is partly composed of cerebral vascular endothelial cells and the tight junctions holding them together. A growing number of studies reveal that PPARγ is expressed in the cerebral vascular endothelium and plays important roles in the regulation of cerebrovascular structure and function (Culman et al., 2007; Beyer et al., 2008; Halabi et al., 2008; Hamblin et al., 2009). For example, PPARγ activation by TZDs can regulate endothelial migration and angiogenesis (Duan et al., 2008), consistent with a role for PPARγ in BBB repair. PPARγ promotes human umbilical vein cell migration by inducing the expression of a member of the class-3 semaphorins, Sema3g (Ming et al., 2015). Activated endothelial cells trigger immune responses through the secretion of pro-inflammatory cytokines/chemokines, such as IL-1 and TNF-α (Ross, 1993, 1999; Tesfamariam and DeFelice, 2007). Many studies have demonstrated that PPARγ agonists reduce the activation and inflammation of endothelial cells by inhibiting proinflammatory cytokines/chemokines (Jackson et al., 1999; Marx et al., 2000; Pasceri et al., 2000; Imamoto et al., 2004; Verrier et al., 2004; Sasaki et al., 2005; Chen et al., 2015). PPARγ in endothelial cells protects against IL-1β-induced endothelial dysfunction (Mukohda et al., 2016). Furthermore, Yin and colleagues found that the PPARγ agonist PGZ significantly reduces OGD-induced endothelial cell death (Yin et al., 2013). In the same study, the effects of adenovirus-mediated gain or loss of PPARγ function strongly support a cytoprotective role for PPARγ in endothelial cultures. Consistent with these in vitro findings, PPARγ agonists also significantly reduce ischemia-triggered increases in cerebrovascular/BBB permeability in mice (Yin et al., 2013). Hind and colleagues established an in vitro BBB model using human brain microvascular endothelial cells and human astrocyte co-cultures (Hind et al., 2015). They discovered that the PPARγ agonist cannabidiol decreased BBB permeability when administered either before or after OGD. Furthermore, vascular cell adhesion molecule-1 expression was decreased. Min et al. reported that activation of PPARγ by telmisartan attenuates BBB impairment in a type 2 diabetic mouse model (Min et al., 2012). In that study, significant decreases in the expression of tight junction proteins (ZO-1, occludin, claudin3, and claudin5) and increases in MMP-2 and MMP-9, oxidative stress, and pro-inflammatory cytokines (MCP-1, IL-6, TNF-α) were observed in the mouse brain after treatment with the PPARα antagonist (Min et al., 2012). Collectively, these studies indicate that PPARγ activation regulates BBB permeability and this may partly underlie its protective role against CNS injuries.

5. PPARγ promotes CNS repair

The chronic phase of CNS injury can progress for weeks to years, even after the CNS commences repair to restore homeostasis. Thus, the injury and reparatory phases are not completely separable and exhibit considerable overlap. During the chronic phase, neural inflammation is eventually controlled and resolved. Debris in and around the lesion zone is isolated and removed by local microglia and/or by infiltrating peripheral immune cells. Neurons may be replenished in limited numbers through neurogenesis. White matter is repaired by neurite outgrowth and remyelination. Angiogenesis and revascularization reseal the compromised BBB. In the next section, we will discuss repair processes, mainly in the chronic phase, and describe the involvement of PPARγ. As discussed in Section 2.4, the existence of PPREs on gene clusters that control cell survival, differentiation, chromatin remodeling, and the DNA damage response support an important role of PPARγ in tissue repair (Lemay and Hwang, 2006; Heinaniemi et al., 2007).

5.1 Resolution of inflammation

5.1.1 An introduction on the resolution of inflammation following CNS injury

Resolution of inflammation is initiated immediately after the onset of CNS challenges, at the same time as the inflammation commences. Clearance of debris is the first step toward the resolution of inflammation, and will be discussed in Section 5.2. Contraction of the immune cell population in the CNS is also essential to restore homeostasis. Neurotrophic factors from the immune system help regenerate injured tissue and prevent cell loss. Anti-inflammatory cells and factors work in concert to complete the process of inflammation resolution.

Both innate and adaptive immune systems have components that regulate inflammation. Immediately after CNS injury, the pro-resolution M2 phenotype predominates in the microglial and macrophage populations for the first 3-5 days, and this is followed by M1 polarization (Hu et al., 2012; Wang et al., 2013). However, within weeks following CNS injury, the pro-inflammatory M1 microglial phenotype gradually subsides and is replaced by the pro-resolution M2 phenotype. The M2 phenotype promotes 1) phagocytosis to clear debris, 2) secretion of anti-inflammatory cytokines, such as IL-10 and transforming growth factor (TGF) β, and 3) release of a plethora of trophic factors, including IGF-1. Concomitantly, infiltrating neutrophils and macrophages undergo apoptosis to help resolve inflammation. The N2 neutrophil population expresses pro-resolution markers, such as CD206 and Ym1, and this is associated with increased neutrophil clearance in the MCAO mouse model (Cuartero et al., 2013). In the adaptive immune system, the best-studied regulatory lymphocytes are known as CD4+CD25+FoxP3+ regulatory T cells (Tregs), which facilitate the resolution of inflammation after CNS injury (Shevach et al., 2006; Zhou et al., 2017). Treg cells can directly suppress effector T cells and myeloid cells by secreting suppressor cytokines such as IL-10, IL-35, and TGF-β, and compete for IL-2, which induces T cell apoptosis due to deprivation of activating cytokines and granzyme-dependent cytotoxicity. Furthermore, by suppressing the functions of antigen presenting cells, Treg cells indirectly modulate the responder T cells. Tregs prevent activation, maturation, and access to effector cells, impeding the immune stimulatory function of antigen presenting cells (Shevach, 2009). In addition, Tregs can express neurotrophic factors such as BDNF and the levels of circulating BDNF+ Tregs are correlated with positive disease outcomes in ischemic stroke patients (Feddersen and Van Ness, 1989).

5.1.2 PPARγ promotes resolution of inflammation

Although beneficial effects of PPARγ agonists have been widely reported in ischemic stroke, ICH, TBI, and SCI, the mechanisms whereby PPARγ promotes resolution of inflammation in acute CNS diseases are poorly understood. Our previous study reported that application of RGZ in a transient MCAO mouse model increased the expression of CD206 on microglia, a marker of the inflammation-resolving M2 phenotype (Han et al., 2015). Furthermore, treatment with RGZ enhances the phagocytic activity of microglia in the mouse distal permanent MCAO model (Ballesteros et al., 2014). Zhao and colleagues reported that RGZ administration promotes hematoma resolution by enhancing microglial phagocytosis after intrastriatal blood injection in both mice and rats (Zhao et al., 2007). Administration of RGZ also promotes infiltration of neutrophils into the ischemic lesion zone, followed by direct neutrophil polarization to the N2 phenotype and microglia/macrophage clearance of neutrophils in the MCAO mouse model (Cuartero et al., 2013). RGZ treatment in mice subjected to permanent MCAO accelerates neutrophil recruitment into the ischemic core, as shown by increased numbers of neutrophils in the ipsilesional hemisphere at 24h after ischemia. RGZ also increased the expression of Ym1 and CD206 by two-fold in these infiltrated neutrophils. Furthermore, RGZ treatment facilitated neutrophil clearance in the infarct core by microglia/macrophages. Taken together, these findings indicate that PPARγ activation might enhance the beneficial functions of microglia and neutrophils. Furthermore, PPARγ agonists prevent excessive activation of neutrophils by promoting their clearance (Cuartero et al., 2013). These collective actions of PPARγ are expected to accelerate tissue repair and functional recovery.

In contrast to the effects of PPARγ activation on microglia and neutrophils, the effects of PPARγ on Tregs in acute CNS diseases are poorly understood. PPARγ is known to be essential for the development of Tregs in highly enriched mouse adipose tissue (Cipolletta et al., 2012; Park and Pan, 2015). Administration of CGZ enhances Treg development and its suppression of Th17 cells in a graft-versus-host disease mouse model (Wohlfert et al., 2007; Park and Pan, 2015). Although the cellular source has not been established, intraperitoneal administration of RGZ after SCI increases the levels of NGF and BDNF (Meng et al., 2011; Mandrekar-Colucci et al., 2013). Furthermore, Tregs can be a source of BDNF and other trophic factors, as discussed in Section 5.1.1.

In conclusion, resolution of inflammation is not an isolated process; it is fully integrated into the chronic recovery phase. Timely clearance of necrotic debris or injured cells prevents oxidative stress and the release of pro-inflammatory factors, impeding the feed-forward cycles that characterize inflammation. Contraction of the immune cell populations after fulfillment of their beneficial functions decreases the risk of excessive immune reactions and the resulting deleterious auto-immune responses. Neurotrophic factors are also essential for the reconstruction of tissue in acute CNS diseases. Direct and indirect evidence outlined above suggests that activation of PPARγ fosters the resolution of inflammation and the recovery of function through some of these mechanisms.

5.2 Clearance of debris and infiltrating cells

5.2.1 An introduction on the clearance of debris and infiltrating cells

Severe CNS injuries cause cell death and the subsequent production of cellular debris. Necrotic or apoptotic neuron somata and axonal debris or detached myelin pieces are detrimental for several reasons. First, they activate immune responses and lead to subsequent chronic inflammation, which causes further cell death and tissue damage. Second, the debris-induced chronic inflammation impairs axonal regeneration and maturation of OPCs, which are vital for post-injury remyelination. Furthermore, infiltrating polymorphonuclear neutrophils (PMNs) and erythrocytes may also be detrimental after hemorrhage and trauma. Application of PMNs onto post-OGD organotypic hippocampal slice cultures causes a remarkable exacerbation of neuronal damage (Neumann et al., 2008), perhaps due to the release of ROS, proteases, and proinflammatory cytokines such as TNF-α (Jordan et al., 1999). In ICH, extravasated erythrocytes in the hematoma undergo lysis within hours to days after ICH, releasing cytotoxic hemoglobin, heme, and iron, thereby negatively influencing the viability of surrounding neurons and oligodendrocytes (Zhao et al., 2009a). Thus, efficient clearance of damaged cellular components by phagocytosis relieves secondary tissue damage and favors tissue repair after CNS injury.

Phagocytosis after CNS injuries is mainly carried out by resident microglia and infiltrated hematogenous macrophages. Using green fluorescent protein (GFP) transgenic bone marrow chimeric mice to distinguish microglia from peripheral macrophages, Schilling and colleagues reported that activated microglia exhibited phagocytic responses beginning on the first day after 30 min of transient MCAO (Schilling et al., 2005). Furthermore, the number of activated microglia increased gradually and peaked on day 10. Peripheral macrophage infiltration was not evident until day 4 after MCAO, peaking in the following 3 days, and then decreasing until 2 weeks after the ischemic insult. Phagocytic microglia accounted for one quarter of total activated microglia and reached maximal levels as early as 24 h post-ischemia and were maintained at this level until the infiltration of hematogenous macrophages. These findings lead to the hypothesis that microglia play a predominant role in debris clearance in the early stages, while assistance from infiltrated macrophages occurs subsequently. Similarly, in a moderate contusion injury model of SCI, Greenhalgh et al. used lysozyme M enhanced GFP-knockin mice to tag hematogenous macrophages and discovered that 1) phagocytic microglia appeared as early as 24 h post-SCI, 2) microglia were the predominant phagocytic cell type until day 3, and 3) infiltrating macrophages become the predominant phagocytic cell on day 3 and persisted for up to 42 days (Greenhalgh and David, 2014). It is noteworthy that only a small percentage of microglia contained phagocytic material at 7d (~3%), while phagocytic material was still evident in a high percentage of infiltrating macrophages at 42 d (~30%) after SCI. Phagocytosis of red blood cells by microglia has also been observed in ICH and promotes hematoma resolution (Zhao et al., 2007). In a closed skull injury-induced mouse TBI model, activated microglia were immunoreactive for galactose-specific lectin (Galectin)-3 (formerly called Mac-2), a receptor involved in the activation of myelin phagocytosis (Rotshenker, 2009). The microglia were most abundant one day following injury, followed by a decrease up to 28 days after TBI, but still were significantly elevated in TBI brains compared to sham control brains (Venkatesan et al., 2010).

The benefits of phagocytosis after acute CNS injuries have been well documented. As mentioned above, external application of invading PMNs to OGD-treated organotypic brain slices dramatically enhances ischemic neurotoxicity (Neumann et al., 2008). However, additional application of microglia counteracted this effect by rapid engulfment of PMNs, followed by neuroprotection that could be abrogated by interfering with microglial PMN engulfment (Neumann et al., 2008). Phagocytosis of blood components by resident microglia and macrophages in ICH mitigates further damage (Zhao et al., 2009a). Accordingly, phagocytosis and subsequent hematoma resolution are associated with improvements in neurological recovery (Chen et al., 2017), whereas loss of phagocytosis exacerbates brain swelling and impedes clot resolution (Ni et al., 2016). Enhanced microglial phagocytosis of myelin and cell debris is also beneficial for the survival of injured neurons and myelin regeneration in acute SCI and TBI, thereby facilitating functional recovery (Rotshenker, 2003; Boekhoff et al., 2012; Redondo-Castro et al., 2013). Molecular mechanisms involved in microglia/macrophage-afforded phagocytosis have been widely studied. In this context, priming is defined as any condition or event that facilitates the interaction between phagocytic receptors and their targets. Priming allows the phagocyte to engulf its targets by exposing ligand binding sites. This exposure endows phagocytic receptors with increased mobility in the plane of the membrane, thereby enhancing the probability of encountering target particles (Freeman and Grinstein, 2014). A large number of inflammatory stimuli promote the transition of the integrins from a bent (closed) conformation to an extended (open) conformation, resulting in enhanced exposure of binding sites (Shattil et al., 2010). Other priming factors include G protein-coupled receptor ligands (Freeman and Grinstein, 2014), Toll-like receptor (TLR) ligands (Patel and Harrison, 2008), and growth factors such as macrophage colony-stimulating factor (Kheir et al., 2005). After priming, specific membrane receptors on phagocytes and downstream signaling pathways contribute to the recognition and engulfment of target microparticles. In general, there are two functionally distinct types of receptors—one with a high binding affinity to foreign microbial pathogens, such as TLRs, and one that recognizes apoptotic cellular substances, such as triggering receptor expressed on myeloid cells 2. During post-injury debris clearance, the latter receptor is more instrumental. Other receptors involved in phagocytosis include Fc receptors (Anderson et al., 1990), complement receptors (Ross et al., 1992), scavenger receptors (Patel et al., 2004), pyrimidinergic receptor P2Y (Koizumi et al., 2007), G-protein coupled receptors (Riyahi et al., 2011; Dustin, 2016) macrophage antigen complex 2 (Rotshenker, 2009), and mannose receptors (Ezekowitz et al., 1990). Furthermore, different surface receptors on phagocytes may be involved in tethering (recognition and binding of cell corpses) and tickling (internalization and activation of downstream signaling) processes. Phosphatidylserine is a dominant “eat me” signal when cells undergo apoptosis (Fadok et al., 1998). The recognition of phosphatidylserine is mediated by at least one tethering and one internalization receptor (Henson and Hume, 2006). For example, integrins and CD36 are involved in tethering, while Gas and protein S are involved in tickling (Majai et al., 2007). Tethering and tickling lead to activation of Rac pathway, which mediates the uptake of dead cells (Majai et al., 2007).

In the context of acute CNS injuries, CD36 and Galectin-3 are the most important and widely studied phagocytosis receptors. CD36, a member of the scavenger receptors, is involved in the recognition and removal of modified lipoproteins, apoptotic cells, and pathogens (Canton et al., 2013). Upregulation of CD36 during the acute (3d) and recovery (7d) periods has been documented in mouse models of ischemia and is correlated with higher phagocytic indices in brain immune cells (Woo et al., 2016). Furthermore, CD36 deficiency compromises microglial phagocytosis of RBCs and slows hematoma absorption (Fang et al., 2014). The importance of CD36 is confirmed by the observation that addition of anti-CD36 antibodies to human macrophages reduces phagocytosis of apoptotic neutrophils by 50% (Fadok et al., 1998). In addition, CD36 is involved both in tethering and ligand internalization processes. Specifically, CD36 functions as an integrin-associated protein and cooperates with integrins to accomplish tethering (Majai et al., 2007). During ligand internalization, CD36 forms a heteromeric complex with β1 and/or β2 integrins and the tetraspanins CD9 and/or CD81, thereby linking to the adaptor Fc receptor γ, which bears an immunoreceptor tyrosine activation motif. By coupling to Fc receptor γ, CD36 is able to engage the Src-family kinases and Syk, which in turn drives the internalization of CD36 and its bound ligands (Majai et al., 2007). Finally, Galectin-3 may activate phagocytosis by upregulating and prolonging KRas-GTP-dependent phosphatidylinositol 3-kinase activity (Rotshenker, 2009).

During phagocytosis, high levels of pro-oxidative molecules and pro-inflammatory mediators are produced and may adversely affect the survival of phagocytes (Zhao et al., 2007; Yi et al., 2008). Thus, resolution of inflammation and oxidative burden facilitate phagocytosis by preventing injury to microglia/macrophages. For example, upregulation of the antioxidant enzyme catalase is known to promote phagocytosis (Donnan et al., 2008).

5.2.2 PPARγ promotes clearance of debris and infiltrating cells

The functional role of PPARγ in microglia/macrophage-afforded phagocytosis has been widely studied. PPARγ is dramatically upregulated during the maturation of human macrophages from monocytes, a process that endows macrophages with more efficient phagocytic capacities (Majai et al., 2007). PPARγ agonists can upregulate microglial phagocytosis of Staphylococcus aureus in bacterial brain abscesses (Kielian et al., 2008). In the brains of Alzheimer’s patients, PPARγ activation is associated with elimination of Aβ by microglia and a reduction in the oxidative burden in damaged tissue (Yamanaka et al., 2012). Consistent with these findings, PPARγ deficiency in macrophages delays skin wound healing in mice by impairing apoptotic cell clearance (Chen et al., 2015). Furthermore, the application of PPARγ antagonists during the differentiation process impairs macrophage-mediated phagocytosis of apoptotic cells, which may be partly attributed to downregulation of CD36 (Majai et al., 2007).

PPARγ-afforded promotion of phagocytosis may be attributed to three factors. First, PPARγ may promote inflammation-resolving M2 polarization in microglia/macrophages. Although somewhat controversial, phagocytosis is more often considered an M2 characteristic (Tang and Le, 2016). Second, PPARγ favors the resolution of inflammation and may therefore reduce proinflammatory cytokines such as IFN-γ, IL-1β, and TNF-α, which could impair the phagocytic activity of microglia (Koenigsknecht-Talboo and Landreth, 2005). Third, PPARγ reduces oxidative stress, which also serves to promote phagocytosis (Donnan et al., 2008).

A number of studies support the view that PPAR-γ activation in microglia and macrophages promotes phagocytosis after CNS injuries. In models of ICH, the activation of PPARγ increases the clearance of blood deposited in primary microglia cultures and in animals, thereby improving hematoma resolution (Heppner et al., 2015). PPARγ activation is followed by upregulation of catalase and CD36 by phagocytes (Donnan et al., 2008) and by inhibition of NF-κB in ICH, all of which serve to boost phagocytosis. Similarly, PPARγ activation with RGZ induces CD36 expression in resident microglia in a mouse model of permanent focal cerebral ischemia, increasing the ability of microglia to engulf dead neutrophils (Ballesteros et al., 2014). The effects of PPARγ activation on phagocytosis in SCI or TBI are not known.

5.3 Neurogenesis

5.3.1 An introduction on neurogenesis in the adult brain after CNS injury

For many years, it was believed that the brain is extremely limited in its capacity for self-renewal (Streilein, 1995). However, we now know that the adult CNS undergoes active self-repair and neurogenesis after CNS injuries, a finding that holds immense translational potential. Two neurogenic niches containing neural stem/progenitor cells (NSCs/NPCs) have been identified in the adult brain—the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampal dentate gyrus (Lindvall and Kokaia, 2015). Neurogenesis in these two niches follows different patterns. Putative NSCs within the SVZ are known as type B cells and are intermixed with ependymal cells. They are characterized by the astrocytic marker glial fibrillary acidic protein (GFAP) and astrocyte-like processes. Once quiescent type B cells are activated, they proliferate and begin to express NPC markers such as nestin. At this stage, they are called transit amplifying precursors or type C cells. After three proliferative cycles, type C cells transform into neuroblasts (type A cells) that migrate towards the olfactory bulb along the rostral migratory stream and differentiate into mature neurons while integrating into olfactory circuits (Doetsch et al., 1997; Quinones-Hinojosa et al., 2006). Neurogenesis in the SGZ originates in another type of NSCs (type I cells) that share properties with radial glial cells and express GFAP and nestin. When activated, type I cells transform into NPCs (type IIa cells) that further develop into type IIb cells and express markers of neuroblasts such as doublecortin. Type IIb cells then generate type III neuroblasts and develop dendrites. Notably, approximately 80% of type III cells die of apoptosis within 2 weeks (Arvidsson et al., 2002), and those that survive mature into NeuN-expressing neurons (Eriksson et al., 1998; Seri et al., 2001). These newly generated neurons form synaptic connections with pyramidal cells of the hippocampus, which play a critical role in cognitive recovery after CNS injury (Seri et al., 2001; Ramirez-Amaya et al., 2006; Clelland et al., 2009; Sahay et al., 2011).

During the past two decades, novel neurogenic niches other than in the SVZ and SGZ have been reported, including in the hypothalamus (Kokoeva et al., 2005; Xu et al., 2005; Migaud et al., 2010; Pierce and Xu, 2010), striatum, neocortical areas, amygdala (Bernier et al., 2002; Dayer et al., 2005; Luzzati et al., 2006), and brainstem (Bauer et al., 2005). Neurogenesis in the spinal cord after SCI is more elusive. Spinal cord ependymal cells may act as NSCs (Duan et al., 2016), but develop into oligodendrocytes and astrocytes and not into neurons, although their NSC potential is preserved in vitro (Johansson et al., 1999; Meletis et al., 2008; Barnabe-Heider et al., 2010). One explanation is that the spinal cord provides a non-permissive microenvironment that specifically blocks neuronal lineage differentiation, unlike the brain (Lee-Liu et al., 2013). However, these ideas await confirmation.

Stroke and TBI both trigger the proliferation, differentiation, and migration of neurons in the SVZ and SGZ (Arvidsson et al., 2002; Masuda et al., 2007; Shen et al., 2008), suggesting a link between CNS injury and neurogenesis. However, whether neurogenesis is vital for CNS recovery after injury is not clear. When neurogenesis is abolished by ablation of doublecortin-expressing neuroblasts in mice subjected to MCAO, infarct sizes and behavioral deficits are increased at 24 h post-injury (Jin et al., 2010a). Furthermore, these effects last up to 12 weeks after MCAO, suggesting a crucial role for neurogenesis in long-term recovery (Wang et al., 2012a). A similar phenomenon is also evident in models of TBI with selective ablation of nestin-expressing NPCs (Blaiss et al., 2011).

5.3.2 PPARγ in neurogenesis

Only a few studies have focused on the role of PPARγ in neurogenesis, and none of them involve CNS injuries. Nevertheless, PPARγ enables normal brain development and global knockout is embryonically lethal (Wada et al., 2006). This has led to the speculation that PPARγ is indispensable in adult neurogenesis. In this section, we will discuss PPARγ-dependent neurogenesis in physiological and disease conditions, and make some inferences about its neurogenic role in CNS injuries. A role for PPARγ in neurogenesis is consistent with genomic studies confirming the presence of PPREs in gene clusters that control cell survival and differentiation (Lemay and Hwang, 2006; Heinaniemi et al., 2007).

PPARγ is essential in normal stem cell proliferation and neural differentiation in vitro. Wada et al. reported a decrease in cell growth rate in cultured PPARγ+/− and PPARγ−/− NSCs (Wada et al., 2006). In PPARγ+/+ NSCs, cell proliferation was enhanced by RGZ and reduced by GW9662. PPARγ agonists can also promote neural differentiation. For example, 15d-PGJ2 promotes the differentiation of embryonic midbrain cells into dopaminergic neurons (Park et al., 2004b). Similarly, PGZ promotes neural differentiation and neurite outgrowth in SH-SY5Y neuroblastoma cells (Park et al., 2004b). Taheri et al. reported an increase in PPARγ mRNA in both the NPC stage and neural cell stage during the differentiation of cultured human embryonic stem cells (hESCs) (Taheri et al., 2015). Notably, NPC markers are decreased by GW9662, but not increased by 15d-PGJ2.

The effects of PPARγ on neurogenesis have been examined in some disease models. During an exploration of hyperglycemia-induced decreases in neurogenesis, advanced glycan end products (AGEs) were found to inhibit NSC proliferation and differentiation, and this was associated with decreased PPARγ expression (Wang et al., 2009). Exogenous RGZ administration rescued AGE-induced repression of NSC proliferation by inhibition of caspase-dependent NSC apoptosis. However, RGZ failed to prevent AGE-mediated inhibition of NSC differentiation (Wang et al., 2009). Intraperitoneal delivery of LPS (5 mg/kg) decreases hippocampal neurogenesis by 50% and this is associated with memory impairments that are reversed by oral intake of RGZ, indicating a protective role of PPARγ against inflammation-induced suppression of neurogenesis (Ormerod et al., 2013). However, conflicting results were reported during congenital cytomegalovirus (CMV) infection of the CNS (Rolland et al., 2016). In that study, CMV infection was associated with increased PPARγ expression and activity, as well as increases in 9-hydroxyoctadecadienoic acid, a known PPARγ agonist. PPARγ activation with 15d-PGJ2 led to impaired neurogenesis in uninfected NSCs, and treatment of NSCs with 9-hydroxyoctadecadienoic acid decreased NSC neural differentiation. Consistent with these findings, treatment of CMV-infected NSCs with the PPARγ inhibitor T0070907 restored normal differentiation rather dramatically. These results suggest complex effects of PPARγ on neurogenesis, and possibly distinct roles in various diseases and experimental models. Although PPARγ is significantly increased by CNS injuries such as cerebrovascular events and trauma, its effects on neurogenic processes remain unknown and need further exploration.

5.4 Remyelination

5.4.1 An introduction on remyelination during CNS repair

Remyelination refers to the progressive restoration of the entire myelin sheath around demyelinated axons, a process that reinstates saltatory conduction (Smith et al., 1979) and resolves functional deficits (Jeffery and Blakemore, 1997; Liebetanz and Merkler, 2006). Remyelination is characterized by the formation of thinner myelin with shorter internodes compared to sheaths that are not injured (Blakemore, 1974). The g ratio refers to the circumference of the axon divided by the circumference of the myelin sheath, and is used to assess the degree of remyelination (Franklin and Ffrench-Constant, 2008). Generally, remyelination consists of three phases: OPC proliferation, OPC recruitment, and OPC maturation. There is consensus that OPCs serve as the major origin of newly generated oligodendrocytes post-demyelination (Dawson et al., 2003). First, retroviral and autoradiographic tracing studies indicate that remyelinating oligodendrocytes originate from dividing cells in normal adult white matter, which are likely to be adult OPCs, although this awaits confirmation (Gensert and Goldman, 1997; Carroll et al., 1998). Second, transplanted OPCs remyelinate demyelinated axons quite effectively (Groves et al., 1993). OPCs repopulate focal demyelination sites, where both oligodendrocytes and OPCs have died, prior to the appearance of new oligodendrocytes; these temporal and spatial patterns strongly suggest that OPCs are indeed the source of remyelinating cells (Sim et al., 2002). Fourth, cells transiently express OPC as well as oligodendrocyte markers at the onset of remyelination (Fancy et al., 2004). In response to injury, microglia, astrocytes, and other inflammatory cells become activated and produce mitogenic factors and pro-migratory factors, including platelet-derived growth factor and basic fibroblast growth factor, which switch OPCs from a quiescent state to a regenerative phenotype and recruit OPCs to the demyelination zones (Franklin and Ffrench-Constant, 2008). In the final phase of remyelination, the recruited OPCs differentiate into myelinating oligodendrocytes and ensheath the demyelinated axons.

The adult brain undergoes remyelination and white matter repair after CNS injuries, but only to a very limited extent. In the closed-skull impact mouse model of TBI, newly generated oligodendrocytes appear as early as 3 days post-injury (Mierzwa et al., 2015). Remyelination is evident within 1 week post-TBI and prolonged for at least 6 weeks. Similar patterns of post-injury remyelination have been reported in animal models of ischemic stroke (Mandai et al., 1997), SCI (Harrison et al., 1975; Tripathi and McTigue, 2007; Hesp et al., 2015), and ICH (Joseph et al., 2016). Remyelination failure can result from failure of OPC recruitment or failure of differentiation. Experimental and clinical data suggest that differentiation is the most vulnerable phase of remyelination (Franklin and Ffrench-Constant, 2008).

5.4.2 PPARγ in remyelination (703)

As mature oligodendrocytic processes form the myelin sheath, promoting NSC differentiation toward the mature oligodendrocytic lineage might directly increase myelination. PPARγ agonists can augment NSC differentiation into mature oligodendrocytes (Kanakasabai et al., 2012; Wan Ibrahim et al., 2013), although the underlying mechanisms remain unknown (Figure 5A).

Figure 5.

Figure 5

PPARγ promotes regeneration and maturation of cells of the oligodendrocyte lineage. (A) PPARγ promotes differentiation of neural stem cells (NSCs) into oligodendrocyte precursor cells (OPCs), and enhances further maturation of OPCs into mature, myelinating oligodendrocytes. (B) Signaling pathways involved in OPC maturation: (1) PPARγ enhances the formation of myelin components (e.g., lipid plasmalogen) by increasing ADAPS expression, which in turn promotes OPC process extension. (2) Rapid lipid metabolism results in oxidative stress and lipid peroxidation, but this is counteracted by PPARγ-mediated upregulation of antioxidants such as catalase and Cu/Zn SOD. (3) PPARγ enhances mitochondrial activation by increasing activity of mitochondrial respiratory chain complex IV and upregulating ATP-induced Ca2+ oscillations. Abbreviations: neural stem cell (NSC), oligodendrocyte precursor cell (OPC), alkyl-dihydroxyacetone phosphate synthase (ADAPS), superoxide dismutase (SOD), reactive oxygen species (ROS), respiratory chain complex I, II, III, and IV, adenosine 5′-triphosphate (ATP), inositol 1,4,5-trisphosphate (IP3), inositol 1,4,5-trisphosphate receptor (IP3R).

The effects of PPARγ in OPC maturation are better studied than its effects on NSCs (Figure 5B). Recent findings demonstrate that PPARγ activation positively contributes to differentiation of multiple lines of OPCs. Studies on B12 glioma-derived cells and primary rat spinal cord OPCs show that PPARγ activation promotes the commitment of OPCs toward differentiation into myelin basic protein-expressing (MBP+) mature oligodendrocytes (Roth et al., 2003). B12 cells express all three PPAR isoforms but only respond to PPARγ agonists. Activation of PPARγ in B12 cells arrests cell proliferation and increases process extension (Roth et al., 2003). Furthermore, treatment with PGZ drives primary rat OPCs to differentiate and mature into oligodendrocytes (Bernardo et al., 2009; De Nuccio et al., 2011). Our recent work demonstrates that RGZ enhances OPC proliferation and increases the number of newly-generated mature oligodendrocytes after MCAO (Han et al., 2015). Nonspecific PPARγ activators such as statins have also been studied for their effects on OPCs. Statins are 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors and activate PPARγ through depletion of geranylgeranyl-pyrophosphate and subsequent RhoA-Rho kinase inactivation (Sim et al., 2008; Paintlia et al., 2010). Statins contribute to activation of PPARγ and directly promote survival and differentiation of rodent and human OPCs in vitro and increase myelin repair in the EAE model of multiple sclerosis in vivo (Paintlia et al., 2005; Miron et al., 2007; Paintlia et al., 2008).

As the ability of PPARγ to promote OPC maturation has been established, the mechanisms underlying this process are also being examined. First, PPARγ is heavily involved in cellular lipid metabolism, and lipid metabolism is of vital importance to oligodendrocyte process formation. PPARγ agonists can promote rat B12 cell maturation by enhancing the formation of myelin-rich lipid plasmalogen, as shown by an increase in alkyl-dihydroxyacetone phosphate synthase (Roth et al., 2003). Second, PPARγ activation blunts the oxidative stress arising from rapid lipid metabolism during OPC differentiation. As mentioned above, recent studies suggest that PPARγ activation upregulates expression of the antioxidant enzymes Cu/Zn SOD and catalase and maintains the equilibrium of the glutathione redox systems, which would mitigate the oxidative stress generated from OPC maturation-related metabolism (Bernardo et al., 2009). Third, PPARγ activation enhances the function of mitochondria during OPC maturation. Repeated administration of PPARγ agonists accelerates primary rat OPC maturation and expression of myelin basic protein by increasing activity of the mitochondrial respiratory chain complex IV and upregulating ADP-induced Ca2+ waves (De Nuccio et al., 2011). In sum, PPARγ appears to facilitate the survival and differentiation of OPC cells and enhance their antioxidant defenses and mitochondrial function (Bernardo et al., 2009).

Despite the abovementioned evidence that PPARγ favors remyelination after injury, negative effects of PPARγ agonists on OPCs have also been observed. For example, 15d-PGJ2 treatment can induce apoptosis in early-stage mouse OPCs (Xiang et al., 2007). Statins can also impede oligodendrocyte maturation and myelin formation in vitro (Miron et al., 2007; Klopfleisch et al., 2008). Statins may also hamper intrinsic myelin repair and maintenance in the cuprizone-induced animal model of demyelination (Klopfleisch et al., 2008; Miron et al., 2009). Furthermore, PPARβ, but not PPARγ, was shown to promote OPC differentiation in primary mixed glial cell cultures enriched in oligodendrocytes (Saluja et al., 2001; Jana et al., 2012). Activation of PPARβ also protects primary human oligodendrocytes against serum deprivation-induced cell death (Jana et al., 2012). However, crosstalk between PPARβ and PPARγ may enhance OPC differentiation in undifferentiated C6 glioma cells (Leisewitz et al., 2008). Therefore, further studies are needed to resolve these controversies and decipher the precise role of PPARγ in regulating developmental processes in cells of the oligodendrocyte lineage. Furthermore, as gene transcriptional activation is a subtle but dynamic process, there needs to be greater resolution of the temporal kinetics of gene expression after PPARγ activation. Other unresolved issues include identification of the stage of OPC differentiation during which agonist-mediated PPARγ activation might be most effective, as well as the pharmacokinetics and optimal dosages of PPARγ agonists.

5.5 BBB repair and angiogenesis/revascularization

Angiogenesis plays important roles in tissue repair and functional recovery after various CNS injuries. Extensive studies have shown that newly formed microvessels improve tissue microperfusion around the ischemia boundary zone and promote functional recovery after stroke (Zhang et al., 2000; Zhang and Chopp, 2002). PPARγ is highly expressed in vascular walls, including the endothelial lining, and is one of major transcription factors involved in the regulation of cerebrovascular structure and function (Culman et al., 2007; Beyer et al., 2008; Halabi et al., 2008; Hamblin et al., 2009). Beyer et al. examined vascular tone and function using heterozygous knock-in mice expressing dominant negative-PPARγ (Beyer et al., 2008). They reported that endothelium-dependent agonist-induced dilation in cerebral blood vessels was significantly impaired in dominant negetive-PPARγ mice. The authors also concluded that interruptions in PPARγ signaling result in endothelial dysfunction through oxidative stress and cause vascular hypertrophy and inward remodeling.

Chu et al. investigated the effects of the PPARγ agonist RGZ on angiogenesis and neurological recovery after cerebral ischemia in the rat MCAO model (Chu et al., 2006). The authors reported that RGZ-pretreated rats present not only with smaller infarct volumes but also with fewer initial neurologic deficits and superior recovery from day 1 through 5 weeks after cerebral ischemia. Notably, RGZ pretreatment also causes significant increases in BrdU+ proliferating endothelial cells in the lesioned hemispheres. Furthermore, RGZ increased cerebral microvessels density, vascular branch points, and vascular surface areas in the ipsilateral hemisphere. The authors conclude that eNOS upregulation may be the main mechanism of RGZ-promoted angiogenesis after focal cerebral ischemia. Another recent study evaluated the role of PPARγ in regulating human pulmonary microvascular endothelial cell migration and angiogenesis (Vattulainen-Collanus et al., 2016). In that study, loss of PPARγ attenuated angiogenesis and migration capacity of the cells through E2F transcription factor 1-mediated Wnt signaling. Using in vivo angiogenesis assays with subcutaneously placed matrigel plugs, Vattulainen-Collanus and colleagues also reported reductions in angiogenesis and mobilization of endothelial progenitor-like cells from the bone marrow in Tie2Cre-PPARγflox/flox mice with ablation of PPARγ in endothelial cells and osteoclasts (Vattulainen-Collanus et al., 2016).

Taken together, these findings suggest that PPARγ activation enhances angiogenesis and functional recovery in rodent ischemic stroke models. The genomic studies discussed in Section 2.4 are consistent with the findings presented here, as they reveal PPREs in genes involved in Wnt signaling, cell growth and maintenance, and BBB integrity (Lemay and Hwang, 2006; Heinaniemi et al., 2007).

6. PPARγ protects against comorbidities

Comorbidities are well known to exacerbate CNS injuries, either by precipitating their onset, or by directly accelerating the underlying pathologic process. Fortunately, many comorbidities, such as hyperglycemia, hypertension, atherosclerosis, and infection are clinically modifiable. Modification of these comorbidities confers protective effects against CNS injury and alleviates disease severity, and is therefore practiced as standard clinical management. Notably, PPARγ serves as a target of many of these comorbid diseases. In this section, we will discuss the effects of modifiable comorbidities commonly associated with CNS injuries, as well as clinical and preclinical evidence favoring their PPARγ-dependent modulation.

6.1 Hyperglycemia

Metabolic syndrome and diabetes mellitus (DM) are known risk factors for stroke. The prevalence of hyperglycemia is 20%–50% in acute stroke, even in the absence of DM (Yu et al., 2015). The second European Cooperative Acute Stroke Study (ECASS-II) followed 748 stroke patients and reported that blood glucose levels were associated with almost all outcomes, including 7-day neurological improvements, 30-day favorable functional outcomes, 90-day negligible dependence, all-cause mortality within 90 days, and hemorrhagic transformation within the first 7 days (Sugawara et al., 2001). Concurrent hyperglycemia is an independent predictor for stroke outcome (Ghajar, 2000; Sarafidis and Lasaridis, 2006). According to the American Stroke Association guidelines, a target of <300 mg/dL blood glucose should be met (Writing Group et al., 2016). Even non-cardiovascular, traumatic CNS injuries such as TBI and SCI are associated with hyperglycemia and severe TBI patients exhibit higher blood glucose levels compared with mild TBI patients (Elder et al., 2004; Jeremitsky et al., 2005). Hyperglycemia is positively correlated with mortality rate, and blood glucose levels are predictive of worse neurological outcomes (Jeremitsky et al., 2005; Uruno et al., 2011; Bevers et al., 2017). Similar detrimental effects of hyperglycemia in SCI have been reported in mouse models as well as in humans (Writing Group et al., 2016). Although impaired neurovascular coupling and reduced cerebral perfusion have been reported in DM patients (Duarte et al., 2015), it is not known with certitude if hyperglycemia is the cause of detrimental effects in the clinic, or if it is a stress response and/or biomarker of injury (Tsai et al., 2009).

TZD treatment is part of the standard of care for DM patients, as it significantly lowers blood glucose levels without elevating the risk of hypoglycemia (Nathan et al., 2009). Not surprisingly, stroke preventive effects of TZD have also been reported. For example, the “Insulin resistance intervention after stroke trial of PGZ” (IRIS trial) (Inzucchi and Furie, 2016) revealed significant secondary stroke prevention, and this effect was more pronounced for the lacunar subtype. It is worth noting that delivery of insulin (and not oral hypoglycemics) is the drug of choice in the acute phase of stroke (Nakayama et al., 1994). A metaanalysis suggests that administration of PPARγ agonists reduces the incidence of primary stroke and recurrent stroke (Liu and Wang, 2015). However, RGZ was reported to increase the risk of heart failure, according to the “Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes” (RECORD) trial (Home et al., 2009), which led to dispensing restrictions by the U.S. Food and Drug Administration (FDA) in 2010. However, after further discussion at the joint meeting of the Endocrinologic and Metabolic Drugs Advisory Committee and the Drug Safety and Risk Management Advisory Committee in 2013 (FDA, 2013), the RECORD trial was deemed to suffer from bias in identification of cardiovascular events and the restrictions were lifted in 2015 (FDA, 2015). There have been no clinical trials on use of PPARγ agonists in TBI or SCI patients thus far.

6.2 Hypertension

Hypertension is the most frequent comorbidity associated with CNS injuries (Abraham et al., 2016; Qureshi and Qureshi, 2017). Hypertension is evident in up to 70% of stroke patients (Staessen et al., 2003). Hypertensive patients experience a two-fold increase in the lifetime risk of stroke compared to subjects with normal blood pressure (BP <120/80 mmHg) (Writing Group et al., 2016). PPARγ has been shown to downregulate BP. Aside from metabolic abnormalities, loss-of-function mutations of PPARγ in humans can lead to severe hypertension (Barroso et al., 1999; Agostini et al., 2006; Auclair et al., 2013). Consistent with these clinical findings, a two-fold increase in PPARγ expression levels can decrease BP by ~2.8 mmHg in rodents (Tsai et al., 2009). The angiotensin II pathway is known to result in hypertension and might be the target of PPARγ. TZDs have been shown to decrease the transcription of angiotensin II receptor type 1 in vascular smooth muscles in vivo (Sugawara et al., 2001), in agreement with in vitro findings that PGZ suppresses angiotensin II-induced aldosterone expression (Uruno et al., 2011). Accordingly, activation of PPARγ with PGZ can attenuate angiotensin II-induced hypertension (Yu et al., 2015). Other mechanisms underlying PPARγ-mediated modulation of BP may include maintaining calcium levels, inhibiting smooth muscle cell proliferation, stabilizing renal function, and attenuating sympathetic over-activation (Sarafidis and Lasaridis, 2006). These findings indicate that PPARγ activation might be a therapeutic approach to lowering BP to prevent the development of stroke. However, BP control during acute stroke is challenging, as both high BP and low BP are independent prognostic factors for poor stroke outcomes (Willmot et al., 2004). Furthermore, in TBI and SCI, intracranial pressure, rather than systemic BP, is one of the most important prognostic factors (Ghajar, 2000), and does not appear to be under direct regulation of systemic BP.

6.3 Dyslipidemia and atherosclerosis

Abnormality in lipid metabolism is an independent risk factor for stroke. Increases in triglycerides are associated with both ischemic stroke and ICH (Nakayama et al., 1994; Ott et al., 1999). In contrast, levels of high-density lipoprotein-cholesterol (HDL-C), a reverse cholesterol transporter, are negatively correlated with stroke risk, and this effect is even more pronounced in thromboembolic stroke (Curb et al., 2004; Huxley et al., 2011). In contrast, low-density lipoprotein (LDL) and peripheral macrophages may directly mediate atherosclerosis (AS) formation. Excessive oxidized LDL deposition induces accumulation of macrophages in the vascular wall. Macrophages engulf detrimental lipoproteins and convert into foam cells (Chistiakov et al., 2016). Activated foam cells within the sclerotic plaques secrete various cytokines, such as IL-6 and TNF-α, which lead to endothelial malfunction (Ait-Oufella et al., 2011), smooth muscle cell migration and proliferation, as well as extracellular matrix production, which may constrict the diameter of the vascular lumen (Wahl et al., 1997). In late stages of AS, excessively narrow arteries limit cerebral blood flow. In addition, macrophages produce extracellular proteases, such as MMPs, which can rupture plaques (Gough et al., 2006), thereby leading to thrombotic stroke (Mughal et al., 2011).

PPARγ expression is high in macrophages within AS plaques (Wilson, 2010). PPARγ may regulate dyslipidemia and AS by modulating macrophage behavior in a number of ways. First, PPARγ promotes clearance of lipid deposition. In apolipoprotein E-knockout mice fed a high-fat diet, TGZ attenuated fatty streak lesion formation and increased HDL-C levels, and this was associated with increased CD36, the scavenger receptor that mediates LDL phagocytosis (Chen et al., 2001). Second, PPARγ prevents excessive engulfment of LDL, thereby reducing macrophage activation. This view is supported by an elegant study showing that, although RGZ cannot inhibit foam-cell transformation induced by LDL in vitro, it can promote cholesterol efflux from macrophages via over-expression of the gene encoding ABCA1 (Chinetti et al., 2001). Third, PPARγ inhibits macrophage-mediated release of inflammatory cytokines in both rodents and humans (Li et al., 2000). PPARγ agonists RGZ and GW7845 strongly inhibit AS development in LDL receptor-deficient mice, and this is associated with decreased TNF-α (Li et al., 2000). Consistent with these findings, RGZ administration in non-diabetic patients was associated with decreased inflammatory markers and adipokines, although no changes in HDL-C and total cholesterol levels were observed (Samaha et al., 2006). Finally, PPARγ mediates growth arrest in vascular smooth muscle cells (Bruemmer et al., 2003a; Bruemmer et al., 2003b), which may serve as another mechanism whereby PPARγ inhibits AS, although this awaits confirmation.

6.4 Renal dysfunction

Renal dysfunction is a common comorbid disease associated with CNS injury and is a risk factor for both ischemic stroke and ICH. Renal dysfunction is apparent in up to one-third of stroke patients and significantly increases mortality (Rowat et al., 2014). In patients with ICH, renal dysfunction is associated with larger lobar hematomas and poorer outcomes (Molshatzki et al., 2011). In TBI, acute renal failure is one of the most common and serious complications, with incidence varying from 1.5% to 18.1% (Schirmer-Mikalsen et al., 2007; Bagshaw et al., 2008). In SCI, renal deterioration was the main cause of death until the introduction of methods that improve bladder emptying in the 1980s (Frankel et al., 1998). SCI-induced renal failure occurs mainly due to increased risk of vesicoureteral reflux and upper urinary tract dilation (McGuire et al., 1981). In a case-control long-term study conducted by Elmelund and colleagues, 26% of SCI patients eventually developed renal deterioration (Elmelund et al., 2017).

A number of studies demonstrate favorable kidney-protective effects of TZDs. Constitutive expression of PPARγ has been detected in human and rodent kidneys, predominantly in the medullary collecting duct (Guan et al., 1997; Yang et al., 1999). In various rodent models of type 2 diabetes, TZD treatment ameliorates diabetic nephropathy and improves renal function, aside from exerting glycemic control (Baylis et al., 2003; Okada et al., 2006). In accordance with preclinical studies, TZD treatment in diabetics significantly decreases urinary albumin excretion in patients with normoalbuminuria, microalbuminuria, or proteinuria (Sarafidis et al., 2010). Other than mitigating diabetic nephropathy, TZDs can also protect against renal dysfunction induced by cyclosporine (Chung et al., 2005), puromycin aminonucleoside (Zuo et al., 2012), aging (Yang et al., 2009a), and polycystic kidney disease (Yoshihara et al., 2011). Mechanisms underlying PPARγ-mediated renal protection include improved insulin resistance and glycemic control, cell-cycle-dependent actions, upregulation of adiponectin expression, anti-inflammatory effects, anti-oxidative effects, prevention of renal lipid accumulation, lowering of BP, etc. (Yang et al., 2009b). However, there are no reports showing that PPARγ protects against renal dysfunction in the setting of CNS injury.

6.5 Infection

Infection is an established risk factor for stroke. Exposure to minor infections in the previous month is an independent risk factor for childhood ischemic stroke, and is evident in one third of cases (Hills et al., 2012). Of these cases, 80% are respiratory infections (Hills et al., 2014). Chronic infections such as periodontitis, chronic bronchitis, and infection with Helicobacter pylori, Chlamydia pneumoniae, and CMV are associated with stroke, although the causal relationship remains unclear (Grau et al., 2010). Infection is also a common complication associated with ischemia, especially in the elderly, and is recognized as an independent predictor of poor outcomes (Wartenberg et al., 2011). Notably, stroke itself may induce immune depression, thereby increasing the incidence of systemic infection (Urra et al., 2009). In addition, infection is associated with excessive ROS generation, which may exacerbate neuronal loss and initiate a self-amplifying cascade of tissue destruction (Berg et al., 2011). Perhaps for this reason, the sepsis death cluster coincides with the so-called “Stroke Belt,” which refers to a region of increased stroke mortality across Mississippi, Alabama, Georgia, Tennessee, Kentucky, North Carolina, and South Carolina (Borhani, 1965; Howard et al., 1995; Wang et al., 2010). This overlap may also suggest shared, unidentified etiology of stroke and sepsis, but this remains to be confirmed.

The effects of PPARγ in sepsis have been examined. PPARγ expression is reduced in the lung in a mouse sepsis model induced by LPS intraperitoneal injection, and 15dPGJ2 improves survival 6-fold (9% to 55%) (Kaplan et al., 2005). These effects are associated with reduced NF-κB and increased HSP70. In another sepsis model induced by cecal ligation and puncture, curcumin treatment increased PPARγ expression in the liver, and reduced mortality and serum TNF-α levels, and these effects were reversed by the PPARγ inhibitor GW9662 (Siddiqui et al., 2006). As far as the underlying mechanism, the PPARγ agonists 15d-PGJ2, BRL 49659, and ciglitazone have been shown to desensitize macrophages in cultures, thereby suppressing the oxidative burst (Von Knethen and Brune, 2001). In addition, 15d-PGJ2 and TGZ can inhibit increased NO production and IκB degradation induced by LPS or heat-killed Escherichia coli and Staphylococcus aureus in rat peritoneal macrophages (Von Knethen and Brune, 2001). Aside from macrophages, T cells are also targeted by PPARγ, as GW9662 inhibits T cell expression of IFN-γ by interfering with c-Jun activation (Cunard et al., 2004).

Whether or not the findings outlined in this section can be translated into CNS injury remains unknown. Furthermore, there exists some controversy, in that PPARγ ligands can induce some proinflammatory responses. For example, macrophagic uptake of oxidized LDL is strongly associated with a proinflammatory phenotype polarization, and this may be enhanced by PPARγ ligands (Tontonoz et al., 1998) (Section 6.3). Second, sepsis refers to a severe infection that disturbs systemic homeostasis. How PPARγ affects mild to moderate infections remains unknown. Third, no reports have focused on the role of PPARγ against infections under conditions of CNS injury. Thus, there remain many gaps in the field.

7. Translational perspectives

A large body of preclinical and clinical work outlined above supports the view that PPARγ activation promotes functional recovery of the CNS by 1) reducing inflammation, 2) minimizing cell death in gray and white matter, and 3) engaging cell repair programs. A wide range of synthetic compounds functioning as PPAR ligands have recently been developed, with TZDs being the most prominent (Lalloyer and Staels, 2010). In addition, it seems likely that endogenous PPARγ activity is naturally increased after acute injuries to help restore homeostasis. Consistent with these views, a clinical study demonstrated that higher plasma concentrations of 15d-PGJ2, a natural PPARγ agonist, were negatively correlated with infarct size and positively correlated with better outcomes in patients with acute athero-thrombotic ischemic stroke (Blanco et al., 2005).

Although there are no clinical reports on the therapeutic effects of PPARγ agonists in the acute stages of cerebral ischemia, the potential effects of PPARγ agonists in stroke prevention and recurrent stroke risk reduction in high-risk populations have been analyzed. We have summarized clinical studies on the therapeutic efficacy and safety of two FDA-approved PPARγ agonists—PGZ and RGZ—in ischemic stroke, and found that most report beneficial outcomes (Table 1). The results of several large-scale clinical trials favor the administration of PPARγ agonists: 1) the prospective PGZ clinical trial in macro-vascular events (PROactive), 2) insulin resistance intervention after stroke trial (IRIS), 3) RGZ evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD), and 4) bypass angioplasty revascularization investigation in type 2 diabetes (BARI2D) (Table 1). Consistent with these observations, a meta-analysis also suggests that administration of PPARγ agonists reduces the incidence of primary and recurrent stroke (Liu and Wang, 2015). Compared with RGZ, PGZ seems to be more favorable, with fewer safety issues and superior therapeutic effects (Liu and Wang, 2015). On the other hand, some of the studies we reviewed failed to show significant effects of PPARγ agonists in stroke prevention (Table 1). Furthermore, RGZ was shown to increase the risk of cardiovascular events and ischemic stroke in some studies, leading to post-marketing monitoring of its safety (FDA, 2015). The adverse effects were mostly found among patients who were elderly or displayed a higher risk of developing macro-vascular diseases (Table 1). In addition, TZDs have been suggested to increase the risk of heart failure, particularly in type 2 diabetics (Michalik and Wahli, 2006). An exacerbation of heart failure following PPARγ activation has also been reported in animal studies (Lygate et al., 2003; Xu et al., 2003). Nevertheless, considering all the clinical studies collectively, it appears that PGZ or RGZ might reduce the incidence of stroke and recurrent stroke, although the target population may need to be carefully selected, as the elderly and those with higher risk of macrovascular diseases might not benefit from such treatment. This type of selective testing of therapies only on the appropriate patient population is gaining acceptance, and future clinical treatments will likely be individualized more carefully based on pharmacogenomic approaches and personalized medicine (Di Sanzo et al., 2017). In addition, BBB permeability and pharmacokinetic-pharmacodynamic properties will need to be carefully considered in order to improve the therapeutic efficacy of PPARγ agonists in CNS diseases. For example, women, diabetic patients, and subjects with lower body weight and higher serum creatinine were found to exhibit lower clearance of the TZD drug rivoglitazone (Rohatagi et al., 2008). For some PPARγ agonists, such as tesaglitazar, rapid absorption, high bioavailability and an elimination half-life of approximately 45 h were observed (Ericsson et al., 2004), suggesting negligible first-pass metabolism. Furthermore, brain penetration of PPARγ agonists such as RGZ may be impaired if they are substrates of P-glycoprotein, a major drug efflux transporter in the BBB (Gold et al., 2010). Given the structural similarities of RGZ and PGZ, the passage of PGZ through the BBB may also be limited (Chang et al., 2015b). Thus, modifications of drug formulation, structure, or dose might be needed to further increase the passage of PPARγ agonists through the BBB. For example, the stereoselectivity of PGZ has been shown to increase its distribution in the CNS and improve its therapeutic efficacy in Alzheimer’s disease (Chang et al., 2015b). Another alternative is to boost the activation of endogenous PPARγ. For example, endogenous prostaglandin J2 activates PPARγ and elicits anti-inflammatory effects (Finch et al., 2017).

Table 1.

Clinical studies of PPARγ agonists safety and therapeutic effects in ischemic stroke.

Study ID Reagent Indication Patient Adverse events Effects Center Ref.
PGZ or RGZ Functional Independence Measure (FIM) of ischemic stroke Stroke with T2DM Not mentioned PGZ and RGZ significantly improved FIM. US (Lee and Reding, 2007)
PGZ or RGZ Incidence of stroke T2DM Heart failure RGZ imposed a higher risk of developing stroke or heart failure, PGZ did not influence risk of stroke China (Lu et al., 2013)
PGZ or RGZ Incidence of stroke T2DM aged 65 or older Not mentioned RGZ increased incidence of stroke. US (Graham et al., 2010)
PGZ or RGZ Incidence of stroke T2DM with high risk of cardiovascular diseases Not mentioned RGZ increased incidence of stroke. US (Shaya et al., 2009)
PGZ or RGZ Incidence of stroke T2DM Not mentioned No significant effects UK (Azoulay et al., 2010)
PGZ or RGZ Incidence of TIA T2DM Not mentioned RGZ increased incidence of TIA Taiwan (Hsiao et al., 2009)
PGZ or RGZ Incidence of stroke and TIA DM Not mentioned No significant effects US (Habib et al., 2009)
PGZ or RGZ Incidence of stroke DM patients aged 65 or older Congestive heart failure No significant effects US (Winkelmayer et al., 2008)
ISRCTNNCT00174993 (PROactive) PGZ Incidenc e of stroke T2DM Edema, heart failure, hypoglycemia, bladder tumors, prostate cancer, weight gain PGZ reduced risk of stroke and recurrent stroke Multicenter (Charbonnel et al., 2004; Dormandy et al., 2005; Katzan, 2007; Wilcox et al., 2007; Erdmann et al., 2010; Scheen, 2012; Erdmann et al., 2014; Erdmann et al., 2016)
NCT00091949 (IRIS) PGZ Incidence of secondary stroke or TIA Insulin-resistant, nondiabetic patients with recent ischemic stroke or TIA Edema, shortness of breath, bone fractures and weight gain PGZ prevented secondary ischemic stroke and myocardial infarction Multicenter (Viscoli et al., 2014; Bath et al., 2016; Inzucchi and Furie, 2016; Kernan et al., 2016)
UMIN000013499 PGZ Incidence of secondary stroke Insulin-resistant, nondiabetic patients with recent ischemic stroke or TIA Edema, rashes, bladder cancer, cytopenia, heart failure PGZ reduced incidence of recurrence of stroke Japan (Tanaka et al., 2015)
PGZ Incidence of stroke T2DM with high risk of cardiovascular diseases Edema, cancer No significant effects Japan (Yoshii et al., 2014)
PGZ Incidence of stroke T2DM Bladder cancer and bone fracture PGZ reduced risk of stroke compared with insulin in T2DM patients US (Vallarino et al., 2013)
RGZ Incidence of stroke Metabolic syndrome undergoing percutaneous coronary intervention (PCI) Not mentioned RGZ displayed a statistical trend to decrease stroke incidence Multicenter (Bhatt et al., 2007)
NCT00379769 (RECORD) RGZ Incidence of stroke T2DM Heart failure, myocardial infarction, bone fracture RGZ reduced incidence of stroke Multicenter (Home et al., 2009; Nissen and Wolski, 2010; Mahaffey et al., 2013)
NCT00006305 (BARI2D) RGZ Incidence of stroke T2DM and coronary artery disease Bone fracture RGZ reduced incidence of stroke Multicenter (Bach et al., 2013)
RGZ Incidence of stroke DM patients aged 65 or older Not mentioned No significant effects Quebec (Vanasse et al., 2009)

8. Summary

PPARγ is a widely-expressed nuclear receptor that regulates the transcription of genes involved in fatty acid storage, glucose homeostasis, insulin sensitivity, anti-inflammatory effects, redox balance, and stem cell differentiation, among other roles. PPARγ regulates PPREs as well as other signaling pathways to elicit neuroprotection and tissue repair. The products of the genes that are modified by PPARγ are found in almost every subcellular compartment as well as the extracellular space. The remarkable breadth of its effects makes PPARγ an attractive therapeutic target for many disorders, including acute brain injury. The activation of Nrf2 and the inhibition of NF-κB are central to the anti-oxidative and anti-inflammatory effects of PPARγ. PPARγ can reduce expression of pro-inflammatory mediators such as IL-6, COX-2, iNOS, and TNF and increase expression of anti-inflammatory mediators such as IL-10 (Chen et al., 2012), which collectively mitigate inflammation in a synergistic fashion. In the acute injury phase, PPARγ may protect against tissue damage by inhibiting neuronal death, demyelination, and BBB disruption. During the chronic recovery phase, PPARγ may facilitate CNS repair by promoting resolution of inflammation, debris clearance, neurogenesis, remyelination, and resealing the BBB. Figure 6 lists the hypothetical mechanisms underlying the protective and reparative effects of PPARγ. Furthermore, PPARγ appears to protect against systemic comorbidities, such as hyperglycemia, hypertension, AS, dyslipidemia, renal dysfunction, and infection, all of which may influence the progression of brain injuries. Many unresolved questions remain, however, including 1) how PPARγ balances the beneficial and destructive faces of neuroinflammation, 2) whether PPARγ directly inhibits demyelination, 3) how PPARγ affects OPC recruitment and differentiation, and 4) whether PPARγ promotes neurogenesis and neuroplasticity. Furthermore, there are serious challenges associated with the application of PPARγ agonists in CNS injury conditions, and some of the effects of PPARγ agonists (such as TZDs) may even be PPARγ-independent, which may cloud the interpretations of preclinical studies. Although preclinical and clinical work supports the therapeutic potential of PPARγ in ischemic stroke, whether PPARγ agonists will be effective in human TBI and SCI is not clear, and this gap in our knowledge is deepened by the poor predictive validity of some of the rodent injury models. Furthermore, PPARγ agonists such as rosiglitazone are associated with weight gain, edema, heart failure, and anemia (Chen et al., 2012). In this respect, however, PPARγ agonists are not unique—there are challenges and limitations of every drug available on the market today, in part due to a failure to account for the genotypic and phenotypic heterogeneity of human populations relative to inbred laboratory animals. At the least, our analysis of the clinical literature in Table 1 and the meta-analyses conducted by Liu and Wang specifically support the use of PPARγ agonists against primary and recurrent stroke in judiciously selected patient populations (Liu and Wang, 2015). Thus, we remain hopeful that identifying the mechanisms underlying the beneficial effects of PPARγ will reveal novel targets for future therapies, and that rational drug design based on PPARγ synthetic or endogenous ligands will lead to superior pharmacological agonists to treat diabetes, cardiovascular diseases, and acute brain disorders.

Figure 6.

Figure 6

Potential mechanisms underlying the CNS protective effects of PPARγ. Abbreviations: antioxidant response element (ARE), blood-brain barrier (BBB), bone marrow (BM), brain-derived neurotrophic factor (BDNF), Cu/Zn superoxide dismutase (Cu/Zn SOD), cyclooxygenase 2 (COX-2), endothelial precursor cells (EPCs), inducible nitric oxide synthase (iNOS), inhibitor of kappa B (IκB), insulin-like growth factor (IGF), matrix metalloproteinases (MMPs), neural stem cell (NSC), nicotinamide adenine dinucleotide phosphate (NADPH), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), oligodendrocyte precursor cell (OPC), reactive oxygen species (ROS), tight junction (TJ), zona occludens (ZO).

Highlights.

  • PPARγ is a master gatekeeper of cell fate decisions

  • PPARγ controls inflammation and oxidative stress and limits acute brain injury

  • PPARγ fosters tissue repair and facilitates long-term functional recovery

  • PPARγ may reduce the risk for stroke in humans

  • PPARγ should be further investigated for its therapeutic potential in acute brain injuries

Acknowledgments

W.C. and T.Y. contributed equally to this article. This work was supported by National Institutes of Health/NINDS grants NS095029 (to J.C., R.K.L., and M.V.L.B.), NS095671 (to J.C.), NS045048 (to J.C.), NS092618 (to X.H.), and R15NS093539 (to R.K.L.), the VA Merit Review awards (I01BX003377 and 1I01BX002495 to J.C.), and grants from the Chinese Natural Science Foundation grants 81529002, 81371306, and 81571285, and (to Y.G. and J.C.). J.C. is a recipient of the VA Senior Research Career Scientist Award, and the Richard King Mellon Endowed Chair and UPMC Endowed Professorship.

Abbreviations

15d-PGJ2

15-deoxy-D12, 15 prostaglandin J2

15(S)-HETE

15(S)-hydroxyeicosatetraenoic acid

ADP

adenosine diphosphate

AGE

advanced glycan end product

AMPA

α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid

ARE

antioxidant response element

AS

atherosclerosis

BBB

blood-brain barrier

BDNF

brain-derived neurotrophic factor

CCI

controlled cortical impact

CDK

cyclin-dependent kinase

CGZ

ciglitazone

CTL

cytotoxic T lymphocyte

CMV

cytomegalovirus

CNS

central nervous system

COX

cyclooxygenase

DC

dendritic cell

DM

diabetes mellitus

EAE

encephalomyelitis

FDA

Food and Drug Administration

Galectin

galactose-specific lectin

GFAP

glial fibrillary acidic protein

GFP

green fluorescent protein

GLT

glutamate transporter

HDAC

histone deacetylase

HDL-C

high-density lipoprotein-cholesterol

HSP

heat shock protein

ICH

intracerebral hemorrhage

IFN

interferon

IGF

insulin-like growth factor

IL

interleukin

iNOS

inducible nitric oxide synthase

LDL

low-density lipoprotein

LPS

lipopolysaccharide

MBP

myelin basic protein

MCAO

middle cerebral artery occlusion

MCP

monocyte chemotactic protein

NCoR

nuclear receptor corepressor

NF

nuclear factor

NGF

nerve growth factor

NMDA

N-methyl-D-aspartic acid

NOX

nicotinamide adenine dinucleotide phosphate oxidase

NPC

neural progenitor cell

NR1C

nuclear receptor 1C

Nrf2

nuclear factor erythroid 2-related factor 2

NSC

neural stem cell

OGD

oxygen-glucose deprivation

OPC

oligodendrocyte precursor cell

PGZ

pioglitazone

PMN

polymorphonuclear neutrophil

PPAR

peroxisome proliferator-activated receptor

PPRE

peroxisome proliferator response element

RGZ

rosiglitazone

ROS

reactive oxygen species

RXR

retinoid X receptor

SAH

subarachnoid hemorrhage

SCI

spinal cord injury

SGZ

subgranular zone

SOD

superoxide dismutase

SVZ

subventricular zone

TAI

traumatic axonal injury

TBI

traumatic brain injury

TGF

transforming growth factor

TGZ

triglitazone

TLR

Toll-like receptor

TNF

tumor necrosis factor

Treg

regulatory T cell

TZD

thiazolidinedione

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abraham HM, Wolfson L, Moscufo N, Guttmann CR, Kaplan RF, White WB. Cardiovascular risk factors and small vessel disease of the brain: Blood pressure, white matter lesions, and functional decline in older persons. J Cereb Blood Flow Metab. 2016;36:132–142. doi: 10.1038/jcbfm.2015.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agostini M, Schoenmakers E, Mitchell C, Szatmari I, Savage D, Smith A, Rajanayagam O, Semple R, Luan J, Bath L, Zalin A, Labib M, Kumar S, Simpson H, Blom D, Marais D, Schwabe J, Barroso I, Trembath R, Wareham N, Nagy L, Gurnell M, O’Rahilly S, Chatterjee K. Non-DNA binding, dominant-negative, human PPARgamma mutations cause lipodystrophic insulin resistance. Cell Metab. 2006;4:303–311. doi: 10.1016/j.cmet.2006.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, Evans RM. PPARgamma signaling and metabolism: the good, the bad and the future. Nat Med. 2013;19:557–566. doi: 10.1038/nm.3159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ait-Oufella H, Taleb S, Mallat Z, Tedgui A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31:969–979. doi: 10.1161/ATVBAHA.110.207415. [DOI] [PubMed] [Google Scholar]
  5. Aktas O, Ullrich O, Infante-Duarte C, Nitsch R, Zipp F. Neuronal damage in brain inflammation. Arch Neurol. 2007;64:185–189. doi: 10.1001/archneur.64.2.185. [DOI] [PubMed] [Google Scholar]
  6. Allahtavakoli M, Moloudi R, Arababadi MK, Shamsizadeh A, Javanmardi K. Delayed post ischemic treatment with Rosiglitazone attenuates infarct volume, neurological deficits and neutrophilia after embolic stroke in rat. Brain Res. 2009;1271:121–127. doi: 10.1016/j.brainres.2009.03.040. [DOI] [PubMed] [Google Scholar]
  7. Anderson CL, Shen L, Eicher DM, Wewers MD, Gill JK. Phagocytosis mediated by three distinct Fc gamma receptor classes on human leukocytes. J Exp Med. 1990;171:1333–1345. doi: 10.1084/jem.171.4.1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Aoun P, Simpkins JW, Agarwal N. Role of PPAR-gamma ligands in neuroprotection against glutamate-induced cytotoxicity in retinal ganglion cells. Invest Ophthalmol Vis Sci. 2003;44:2999–3004. doi: 10.1167/iovs.02-1060. [DOI] [PubMed] [Google Scholar]
  9. Appel S, Mirakaj V, Bringmann A, Weck MM, Grunebach F, Brossart P. PPAR-gamma agonists inhibit toll-like receptor-mediated activation of dendritic cells via the MAP kinase and NF-kappaB pathways. Blood. 2005;106:3888–3894. doi: 10.1182/blood-2004-12-4709. [DOI] [PubMed] [Google Scholar]
  10. Arbo BD, Bennetti F, Ribeiro MF. Astrocytes as a target for neuroprotection: Modulation by progesterone and dehydroepiandrosterone. Prog Neurobiol. 2016;144:27–47. doi: 10.1016/j.pneurobio.2016.03.010. [DOI] [PubMed] [Google Scholar]
  11. Armstrong RC, Mierzwa AJ, Marion CM, Sullivan GM. White matter involvement after TBI: Clues to axon and myelin repair capacity. Exp Neurol. 2016a;275(Pt 3):328–333. doi: 10.1016/j.expneurol.2015.02.011. [DOI] [PubMed] [Google Scholar]
  12. Armstrong RC, Mierzwa AJ, Sullivan GM, Sanchez MA. Myelin and oligodendrocyte lineage cells in white matter pathology and plasticity after traumatic brain injury. Neuropharmacology. 2016b;110:654–659. doi: 10.1016/j.neuropharm.2015.04.029. [DOI] [PubMed] [Google Scholar]
  13. Aronowski J, Zhao X. Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke. 2011;42:1781–1786. doi: 10.1161/STROKEAHA.110.596718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O. Neuronal replacement from endogenous precursors in the adult brain after stroke. Nat Med. 2002;8:963–970. doi: 10.1038/nm747. [DOI] [PubMed] [Google Scholar]
  15. Asano T. Oxyhemoglobin as the principal cause of cerebral vasospasm: a holistic view of its actions. Crit Rev Neurosurg. 1999;9:303–318. doi: 10.1007/s003290050147. [DOI] [PubMed] [Google Scholar]
  16. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015;212:991–999. doi: 10.1084/jem.20142290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Attwell D, Mishra A, Hall CN, O’Farrell FM, Dalkara T. What is a pericyte? J Cereb Blood Flow Metab. 2016;36:451–455. doi: 10.1177/0271678X15610340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Auclair M, Vigouroux C, Boccara F, Capel E, Vigeral C, Guerci B, Lascols O, Capeau J, Caron-Debarle M. Peroxisome proliferator-activated receptor-gamma mutations responsible for lipodystrophy with severe hypertension activate the cellular renin-angiotensin system. Arterioscler Thromb Vasc Biol. 2013;33:829–838. doi: 10.1161/ATVBAHA.112.300962. [DOI] [PubMed] [Google Scholar]
  19. Azoulay L, Schneider-Lindner V, Dell’aniello S, Filion KB, Suissa S. Thiazolidinediones and the risk of incident strokes in patients with type 2 diabetes: a nested case-control study. Pharmacoepidemiol Drug Saf. 2010;19:343–350. doi: 10.1002/pds.1883. [DOI] [PubMed] [Google Scholar]
  20. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL, Investigators, B.D. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation. 2013;128:785–794. doi: 10.1161/CIRCULATIONAHA.112.000678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bagshaw SM, George C, Gibney RT, Bellomo R. A multi-center evaluation of early acute kidney injury in critically ill trauma patients. Ren Fail. 2008;30:581–589. doi: 10.1080/08860220802134649. [DOI] [PubMed] [Google Scholar]
  22. Ballesteros I, Cuartero MI, Pradillo JM, de la Parra J, Perez-Ruiz A, Corbi A, Ricote M, Hamilton JA, Sobrado M, Vivancos J, Nombela F, Lizasoain I, Moro MA. Rosiglitazone-induced CD36 up-regulation resolves inflammation by PPARgamma and 5-LO-dependent pathways. J Leukoc Biol. 2014;95:587–598. doi: 10.1189/jlb.0613326. [DOI] [PubMed] [Google Scholar]
  23. Barnabe-Heider F, Goritz C, Sabelstrom H, Takebayashi H, Pfrieger FW, Meletis K, Frisen J. Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell. 2010;7:470–482. doi: 10.1016/j.stem.2010.07.014. [DOI] [PubMed] [Google Scholar]
  24. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, Maslen GL, Williams TD, Lewis H, Schafer AJ, Chatterjee VK, O’Rahilly S. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature. 1999;402:880–883. doi: 10.1038/47254. [DOI] [PubMed] [Google Scholar]
  25. Bath PM, Appleton JP, Sprigg N. The Insulin Resistance Intervention after Stroke trial: A perspective on future practice and research. Int J Stroke. 2016;11:741–743. doi: 10.1177/1747493016660099. [DOI] [PubMed] [Google Scholar]
  26. Bauer S, Hay M, Amilhon B, Jean A, Moyse E. In vivo neurogenesis in the dorsal vagal complex of the adult rat brainstem. Neuroscience. 2005;130:75–90. doi: 10.1016/j.neuroscience.2004.08.047. [DOI] [PubMed] [Google Scholar]
  27. Baylis C, Atzpodien EA, Freshour G, Engels K. Peroxisome proliferator-activated receptor [gamma] agonist provides superior renal protection versus angiotensin-converting enzyme inhibition in a rat model of type 2 diabetes with obesity. J Pharmacol Exp Ther. 2003;307:854–860. doi: 10.1124/jpet.103.055616. [DOI] [PubMed] [Google Scholar]
  28. Bayly PV, Cohen TS, Leister EP, Ajo D, Leuthardt EC, Genin GM. Deformation of the human brain induced by mild acceleration. J Neurotrauma. 2005;22:845–856. doi: 10.1089/neu.2005.22.845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Benarroch EE. Oligodendrocytes: Susceptibility to injury and involvement in neurologic disease. Neurology. 2009;72:1779–1785. doi: 10.1212/WNL.0b013e3181a6b123. [DOI] [PubMed] [Google Scholar]
  30. Berg RM, Moller K, Bailey DM. Neuro-oxidative-nitrosative stress in sepsis. J Cereb Blood Flow Metab. 2011;31:1532–1544. doi: 10.1038/jcbfm.2011.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med. 2002;53:409–435. doi: 10.1146/annurev.med.53.082901.104018. [DOI] [PubMed] [Google Scholar]
  32. Bernardo A, Bianchi D, Magnaghi V, Minghetti L. Peroxisome proliferator-activated receptor-gamma agonists promote differentiation and antioxidant defenses of oligodendrocyte progenitor cells. J Neuropathol Exp Neurol. 2009;68:797–808. doi: 10.1097/NEN.0b013e3181aba2c1. [DOI] [PubMed] [Google Scholar]
  33. Bernardo A, Levi G, Minghetti L. Role of the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) and its natural ligand 15-deoxy-Delta12, 14-prostaglandin J2 in the regulation of microglial functions. Eur J Neurosci. 2000;12:2215–2223. doi: 10.1046/j.1460-9568.2000.00110.x. [DOI] [PubMed] [Google Scholar]
  34. Bernier PJ, Bedard A, Vinet J, Levesque M, Parent A. Newly generated neurons in the amygdala and adjoining cortex of adult primates. Proc Natl Acad Sci U S A. 2002;99:11464–11469. doi: 10.1073/pnas.172403999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Bevers MB, Vaishnav NH, Pham L, Battey TW, Kimberly WT. Hyperglycemia is associated with more severe cytotoxic injury after stroke. J Cereb Blood Flow Metab. 2017;37:2577–2583. doi: 10.1177/0271678X16671730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Beyer AM, Baumbach GL, Halabi CM, Modrick ML, Lynch CM, Gerhold TD, Ghoneim SM, de Lange WJ, Keen HL, Tsai YS, Maeda N, Sigmund CD, Faraci FM. Interference with PPARgamma signaling causes cerebral vascular dysfunction, hypertrophy, and remodeling. Hypertension. 2008;51:867–871. doi: 10.1161/HYPERTENSIONAHA.107.103648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Bhatt DL, Chew DP, Grines C, Mukherjee D, Leesar M, Gilchrist IC, Corbelli JC, Blankenship JC, Eres A, Steinhubl S, Tan WA, Resar JR, AlMahameed A, Abdel-Latif A, Tang WH, Brennan D, McErlean E, Hazen SL, Topol EJ. Peroxisome proliferator-activated receptor gamma agonists for the Prevention of Adverse events following percutaneous coronary Revascularization–results of the PPAR study. Am Heart J. 2007;154:137–143. doi: 10.1016/j.ahj.2007.03.029. [DOI] [PubMed] [Google Scholar]
  38. Blaiss CA, Yu TS, Zhang G, Chen J, Dimchev G, Parada LF, Powell CM, Kernie SG. Temporally specified genetic ablation of neurogenesis impairs cognitive recovery after traumatic brain injury. J Neurosci. 2011;31:4906–4916. doi: 10.1523/JNEUROSCI.5265-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Blakemore WF. Pattern of remyelination in the CNS. Nature. 1974;249:577–578. doi: 10.1038/249577a0. [DOI] [PubMed] [Google Scholar]
  40. Blanco M, Moro MA, Davalos A, Leira R, Castellanos M, Serena J, Vivancos J, Rodriguez-Yanez M, Lizasoain I, Castillo J. Increased plasma levels of 15-deoxyDelta prostaglandin J2 are associated with good outcome in acute atherothrombotic ischemic stroke. Stroke. 2005;36:1189–1194. doi: 10.1161/01.STR.0000166054.55993.e5. [DOI] [PubMed] [Google Scholar]
  41. Boekhoff TM, Ensinger EM, Carlson R, Bock P, Baumgartner W, Rohn K, Tipold A, Stein VM. Microglial contribution to secondary injury evaluated in a large animal model of human spinal cord trauma. J Neurotrauma. 2012;29:1000–1011. doi: 10.1089/neu.2011.1821. [DOI] [PubMed] [Google Scholar]
  42. Borhani NO. Changes and Geographic Distribution of Mortality from Cerebrovascular Disease. Am J Public Health Nations Health. 1965;55:673–681. doi: 10.2105/ajph.55.5.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Braissant O, Wahli W. Differential expression of peroxisome proliferator-activated receptor-alpha, -beta, and -gamma during rat embryonic development. Endocrinology. 1998;139:2748–2754. doi: 10.1210/endo.139.6.6049. [DOI] [PubMed] [Google Scholar]
  44. Bruemmer D, Berger JP, Liu J, Kintscher U, Wakino S, Fleck E, Moller DE, Law RE. A non-thiazolidinedione partial peroxisome proliferator-activated receptor gamma ligand inhibits vascular smooth muscle cell growth. Eur J Pharmacol. 2003a;466:225–234. doi: 10.1016/s0014-2999(03)01556-5. [DOI] [PubMed] [Google Scholar]
  45. Bruemmer D, Yin F, Liu J, Berger JP, Sakai T, Blaschke F, Fleck E, Van Herle AJ, Forman BM, Law RE. Regulation of the growth arrest and DNA damage-inducible gene 45 (GADD45) by peroxisome proliferator-activated receptor gamma in vascular smooth muscle cells. Circ Res. 2003b;93:e38–47. doi: 10.1161/01.RES.0000088344.15288.E6. [DOI] [PubMed] [Google Scholar]
  46. Butt AM. Neurotransmitter-mediated calcium signalling in oligodendrocyte physiology and pathology. Glia. 2006;54:666–675. doi: 10.1002/glia.20424. [DOI] [PubMed] [Google Scholar]
  47. Canton J, Neculai D, Grinstein S. Scavenger receptors in homeostasis and immunity. Nat Rev Immunol. 2013;13:621–634. doi: 10.1038/nri3515. [DOI] [PubMed] [Google Scholar]
  48. Carroll WM, Jennings AR, Ironside LJ. Identification of the adult resting progenitor cell by autoradiographic tracking of oligodendrocyte precursors in experimental CNS demyelination. Brain. 1998;121(Pt 2):293–302. doi: 10.1093/brain/121.2.293. [DOI] [PubMed] [Google Scholar]
  49. Carson MJ, Doose JM, Melchior B, Schmid CD, Ploix CC. CNS immune privilege: hiding in plain sight. Immunol Rev. 2006;213:48–65. doi: 10.1111/j.1600-065X.2006.00441.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Chaitanya GV, Eeka P, Munker R, Alexander JS, Babu PP. Role of cytotoxic protease granzyme-b in neuronal degeneration during human stroke. Brain Pathol. 2011;21:16–30. doi: 10.1111/j.1750-3639.2010.00426.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Chang CZ, Wu SC, Kwan AL. Glycyrrhizin Attenuates Proinflammatory Cytokines through a Peroxisome Proliferator-Activated Receptor-gamma-Dependent Mechanism and Experimental Vasospasm in a Rat Model. J Vasc Res. 2015a;52:12–21. doi: 10.1159/000381099. [DOI] [PubMed] [Google Scholar]
  52. Chang KL, Pee HN, Yang S, Ho PC. Influence of drug transporters and stereoselectivity on the brain penetration of pioglitazone as a potential medicine against Alzheimer’s disease. Sci Rep. 2015b;5:9000. doi: 10.1038/srep09000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Charbonnel B, Dormandy J, Erdmann E, Massi-Benedetti M, Skene A, Group, P.R.S. The prospective pioglitazone clinical trial in macrovascular events (PROactive): can pioglitazone reduce cardiovascular events in diabetes? Study design and baseline characteristics of 5238 patients. Diabetes Care. 2004;27:1647–1653. doi: 10.2337/diacare.27.7.1647. [DOI] [PubMed] [Google Scholar]
  54. Chen H, Shi R, Luo B, Yang X, Qiu L, Xiong J, Jiang M, Liu Y, Zhang Z, Wu Y. Macrophage peroxisome proliferator-activated receptor gamma deficiency delays skin wound healing through impairing apoptotic cell clearance in mice. Cell Death Dis. 2015;6:e1597. doi: 10.1038/cddis.2014.544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Chen Q, Shi X, Tan Q, Feng Z, Wang Y, Yuan Q, Tao Y, Zhang J, Tan L, Zhu G, Feng H, Chen Z. Simvastatin Promotes Hematoma Absorption and Reduces Hydrocephalus Following Intraventricular Hemorrhage in Part by Upregulating CD36. Transl Stroke Res. 2017 doi: 10.1007/s12975-017-0521-y. [DOI] [PubMed] [Google Scholar]
  56. Chen S, Feng H, Sherchan P, Klebe D, Zhao G, Sun X, Zhang J, Tang J, Zhang JH. Controversies and evolving new mechanisms in subarachnoid hemorrhage. Prog Neurobiol. 2014;115:64–91. doi: 10.1016/j.pneurobio.2013.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Chen YC, Wu JS, Tsai HD, Huang CY, Chen JJ, Sun GY, Lin TN. Peroxisome proliferator-activated receptor gamma (PPAR-gamma) and neurodegenerative disorders. Mol Neurobiol. 2012;46:114–124. doi: 10.1007/s12035-012-8259-8. [DOI] [PubMed] [Google Scholar]
  58. Chen Z, Ishibashi S, Perrey S, Osuga J, Gotoda T, Kitamine T, Tamura Y, Okazaki H, Yahagi N, Iizuka Y, Shionoiri F, Ohashi K, Harada K, Shimano H, Nagai R, Yamada N. Troglitazone inhibits atherosclerosis in apolipoprotein E-knockout mice: pleiotropic effects on CD36 expression and HDL. Arterioscler Thromb Vasc Biol. 2001;21:372–377. doi: 10.1161/01.atv.21.3.372. [DOI] [PubMed] [Google Scholar]
  59. Chinetti G, Lestavel S, Bocher V, Remaley AT, Neve B, Torra IP, Teissier E, Minnich A, Jaye M, Duverger N, Brewer HB, Fruchart JC, Clavey V, Staels B. PPAR-alpha and PPAR-gamma activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat Med. 2001;7:53–58. doi: 10.1038/83348. [DOI] [PubMed] [Google Scholar]
  60. Ching J, Amiridis S, Stylli SS, Bjorksten AR, Kountouri N, Zheng T, Paradiso L, Luwor RB, Morokoff AP, O’Brien TJ, Kaye AH. The peroxisome proliferator activated receptor gamma agonist pioglitazone increases functional expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in human glioblastoma cells. Oncotarget. 2015;6:21301–21314. doi: 10.18632/oncotarget.4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Chistiakov DA, Bobryshev YV, Orekhov AN. Macrophage-mediated cholesterol handling in atherosclerosis. J Cell Mol Med. 2016;20:17–28. doi: 10.1111/jcmm.12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Cho HY, Gladwell W, Wang X, Chorley B, Bell D, Reddy SP, Kleeberger SR. Nrf2-regulated PPAR{gamma} expression is critical to protection against acute lung injury in mice. Am J Respir Crit Care Med. 2010;182:170–182. doi: 10.1164/rccm.200907-1047OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Choi JH, Banks AS, Estall JL, Kajimura S, Bostrom P, Laznik D, Ruas JL, Chalmers MJ, Kamenecka TM, Bluher M, Griffin PR, Spiegelman BM. Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARgamma by Cdk5. Nature. 2010;466:451–456. doi: 10.1038/nature09291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Choi JH, Banks AS, Kamenecka TM, Busby SA, Chalmers MJ, Kumar N, Kuruvilla DS, Shin Y, He Y, Bruning JB, Marciano DP, Cameron MD, Laznik D, Jurczak MJ, Schurer SC, Vidovic D, Shulman GI, Spiegelman BM, Griffin PR. Antidiabetic actions of a non-agonist PPARgamma ligand blocking Cdk5-mediated phosphorylation. Nature. 2011;477:477–481. doi: 10.1038/nature10383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Choi YS, Cho HY, Hoyt KR, Naegele JR, Obrietan K. IGF-1 receptor-mediated ERK/MAPK signaling couples status epilepticus to progenitor cell proliferation in the subgranular layer of the dentate gyrus. Glia. 2008;56:791–800. doi: 10.1002/glia.20653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Chu K, Lee ST, Koo JS, Jung KH, Kim EH, Sinn DI, Kim JM, Ko SY, Kim SJ, Song EC, Kim M, Roh JK. Peroxisome proliferator-activated receptor-gamma-agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia. Brain Res. 2006;1093:208–218. doi: 10.1016/j.brainres.2006.03.114. [DOI] [PubMed] [Google Scholar]
  67. Chung BH, Li C, Sun BK, Lim SW, Ahn KO, Yang JH, Choi YH, Yoon KH, Sugawara A, Ito S, Kim J, Yang CW. Rosiglitazone protects against cyclosporine-induced pancreatic and renal injury in rats. Am J Transplant. 2005;5:1856–1867. doi: 10.1111/j.1600-6143.2005.00979.x. [DOI] [PubMed] [Google Scholar]
  68. Cipolletta D, Feuerer M, Li A, Kamei N, Lee J, Shoelson SE, Benoist C, Mathis D. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature. 2012;486:549–553. doi: 10.1038/nature11132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Puddington L, Padula SJ. The nuclear receptor PPAR gamma and immunoregulation: PPAR gamma mediates inhibition of helper T cell responses. J Immunol. 2000;164:1364–1371. doi: 10.4049/jimmunol.164.3.1364. [DOI] [PubMed] [Google Scholar]
  70. Clark RK, Lee EV, Fish CJ, White RF, Price WJ, Jonak ZL, Feuerstein GZ, Barone FC. Development of tissue damage, inflammation and resolution following stroke: an immunohistochemical and quantitative planimetric study. Brain Res Bull. 1993;31:565–572. doi: 10.1016/0361-9230(93)90124-t. [DOI] [PubMed] [Google Scholar]
  71. Clark RS, Schiding JK, Kaczorowski SL, Marion DW, Kochanek PM. Neutrophil accumulation after traumatic brain injury in rats: comparison of weight drop and controlled cortical impact models. J Neurotrauma. 1994;11:499–506. doi: 10.1089/neu.1994.11.499. [DOI] [PubMed] [Google Scholar]
  72. Clarkson BD, Heninger E, Harris MG, Lee J, Sandor M, Fabry Z. Innate-adaptive crosstalk: how dendritic cells shape immune responses in the CNS. Adv Exp Med Biol. 2012;946:309–333. doi: 10.1007/978-1-4614-0106-3_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Clarner T, Diederichs F, Berger K, Denecke B, Gan L, van der Valk P, Beyer C, Amor S, Kipp M. Myelin debris regulates inflammatory responses in an experimental demyelination animal model and multiple sclerosis lesions. Glia. 2012;60:1468–1480. doi: 10.1002/glia.22367. [DOI] [PubMed] [Google Scholar]
  74. Clelland CD, Choi M, Romberg C, Clemenson GD, Jr, Fragniere A, Tyers P, Jessberger S, Saksida LM, Barker RA, Gage FH, Bussey TJ. A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science. 2009;325:210–213. doi: 10.1126/science.1173215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Collino M, Aragno M, Mastrocola R, Gallicchio M, Rosa AC, Dianzani C, Danni O, Thiemermann C, Fantozzi R. Modulation of the oxidative stress and inflammatory response by PPAR-gamma agonists in the hippocampus of rats exposed to cerebral ischemia/reperfusion. Eur J Pharmacol. 2006;530:70–80. doi: 10.1016/j.ejphar.2005.11.049. [DOI] [PubMed] [Google Scholar]
  76. Compe E, Drane P, Laurent C, Diderich K, Braun C, Hoeijmakers JH, Egly JM. Dysregulation of the peroxisome proliferator-activated receptor target genes by XPD mutations. Mol Cell Biol. 2005;25:6065–6076. doi: 10.1128/MCB.25.14.6065-6076.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Conti AC, Raghupathi R, Trojanowski JQ, McIntosh TK. Experimental brain injury induces regionally distinct apoptosis during the acute and delayed post-traumatic period. J Neurosci. 1998;18:5663–5672. doi: 10.1523/JNEUROSCI.18-15-05663.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Cuartero MI, Ballesteros I, Moraga A, Nombela F, Vivancos J, Hamilton JA, Corbi AL, Lizasoain I, Moro MA. N2 neutrophils, novel players in brain inflammation after stroke: modulation by the PPARgamma agonist rosiglitazone. Stroke. 2013;44:3498–3508. doi: 10.1161/STROKEAHA.113.002470. [DOI] [PubMed] [Google Scholar]
  79. Cullingford TE, Bhakoo K, Peuchen S, Dolphin CT, Patel R, Clark JB. Distribution of mRNAs encoding the peroxisome proliferator-activated receptor alpha, beta, and gamma and the retinoid X receptor alpha, beta, and gamma in rat central nervous system. J Neurochem. 1998;70:1366–1375. doi: 10.1046/j.1471-4159.1998.70041366.x. [DOI] [PubMed] [Google Scholar]
  80. Culman J, Zhao Y, Gohlke P, Herdegen T. PPAR-gamma: therapeutic target for ischemic stroke. Trends Pharmacol Sci. 2007;28:244–249. doi: 10.1016/j.tips.2007.03.004. [DOI] [PubMed] [Google Scholar]
  81. Cunard R, Eto Y, Muljadi JT, Glass CK, Kelly CJ, Ricote M. Repression of IFN-gamma expression by peroxisome proliferator-activated receptor gamma. J Immunol. 2004;172:7530–7536. doi: 10.4049/jimmunol.172.12.7530. [DOI] [PubMed] [Google Scholar]
  82. Curb JD, Abbott RD, Rodriguez BL, Masaki KH, Chen R, Popper JS, Petrovitch H, Ross GW, Schatz IJ, Belleau GC, Yano K. High density lipoprotein cholesterol and the risk of stroke in elderly men: the Honolulu heart program. Am J Epidemiol. 2004;160:150–157. doi: 10.1093/aje/kwh177. [DOI] [PubMed] [Google Scholar]
  83. da Rocha Junior LF, Dantas AT, Duarte AL, de Melo Rego MJ, Pitta Ida R, Pitta MG. PPARgamma Agonists in Adaptive Immunity: What Do Immune Disorders and Their Models Have to Tell Us? PPAR Res. 2013;2013:519724. doi: 10.1155/2013/519724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Dawson MR, Polito A, Levine JM, Reynolds R. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci. 2003;24:476–488. doi: 10.1016/s1044-7431(03)00210-0. [DOI] [PubMed] [Google Scholar]
  85. Dayer AG, Cleaver KM, Abouantoun T, Cameron HA. New GABAergic interneurons in the adult neocortex and striatum are generated from different precursors. J Cell Biol. 2005;168:415–427. doi: 10.1083/jcb.200407053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. De Nuccio C, Bernardo A, De Simone R, Mancuso E, Magnaghi V, Visentin S, Minghetti L. Peroxisome proliferator-activated receptor gamma agonists accelerate oligodendrocyte maturation and influence mitochondrial functions and oscillatory Ca(2+) waves. J Neuropathol Exp Neurol. 2011;70:900–912. doi: 10.1097/NEN.0b013e3182309ab1. [DOI] [PubMed] [Google Scholar]
  87. de Wit NM, Vanmol J, Kamermans A, Hendriks J, de Vries HE. Inflammation at the blood-brain barrier: The role of liver X receptors. Neurobiol Dis. 2017 doi: 10.1016/j.nbd.2016.09.015. [DOI] [PubMed] [Google Scholar]
  88. Dekkers JF, van der Ent CK, Kalkhoven E, Beekman JM. PPARgamma as a therapeutic target in cystic fibrosis. Trends Mol Med. 2012;18:283–291. doi: 10.1016/j.molmed.2012.03.004. [DOI] [PubMed] [Google Scholar]
  89. Delerive P, Furman C, Teissier E, Fruchart J, Duriez P, Staels B. Oxidized phospholipids activate PPARalpha in a phospholipase A2-dependent manner. FEBS Lett. 2000;471:34–38. doi: 10.1016/s0014-5793(00)01364-8. [DOI] [PubMed] [Google Scholar]
  90. Dello Russo C, Gavrilyuk V, Weinberg G, Almeida A, Bolanos JP, Palmer J, Pelligrino D, Galea E, Feinstein DL. Peroxisome proliferator-activated receptor gamma thiazolidinedione agonists increase glucose metabolism in astrocytes. J Biol Chem. 2003;278:5828–5836. doi: 10.1074/jbc.M208132200. [DOI] [PubMed] [Google Scholar]
  91. Desmond DW. Cognition and white matter lesions. Cerebrovasc Dis. 2002;13(Suppl 2):53–57. doi: 10.1159/000049151. [DOI] [PubMed] [Google Scholar]
  92. Dhawan V. Studies on Respiratory Disorders. Springer; 2014. Reactive Oxygen and Nitrogen Species: General Considerations; pp. 27–47. [Google Scholar]
  93. Di Sanzo M, Fineschi V, Borro M, La Russa R, Santurro A, Scopetti M, Simmaco M, Frati P. Clinical applications of Personalized Medicine: a new paradigm and challenge. Curr Pharm Biotechnol. 2017 doi: 10.2174/1389201018666170224105600. [DOI] [PubMed] [Google Scholar]
  94. Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci. 1999;22:391–397. doi: 10.1016/s0166-2236(99)01401-0. [DOI] [PubMed] [Google Scholar]
  95. Doetsch F, Garcia-Verdugo JM, Alvarez-Buylla A. Cellular composition and three-dimensional organization of the subventricular germinal zone in the adult mammalian brain. J Neurosci. 1997;17:5046–5061. doi: 10.1523/JNEUROSCI.17-13-05046.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Donnan GA, Fisher M, Macleod M, Davis SM. Stroke. Lancet. 2008;371:1612–1623. doi: 10.1016/S0140-6736(08)60694-7. [DOI] [PubMed] [Google Scholar]
  97. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J, Investigators, P.R. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet. 2005;366:1279–1289. doi: 10.1016/S0140-6736(05)67528-9. [DOI] [PubMed] [Google Scholar]
  98. Duan H, Song W, Zhao W, Gao Y, Yang Z, Li X. Endogenous neurogenesis in adult mammals after spinal cord injury. Sci China Life Sci. 2016;59:1313–1318. doi: 10.1007/s11427-016-0205-2. [DOI] [PubMed] [Google Scholar]
  99. Duan SZ, Usher MG, Mortensen RM. Peroxisome proliferator-activated receptor-gamma-mediated effects in the vasculature. Circ Res. 2008;102:283–294. doi: 10.1161/CIRCRESAHA.107.164384. [DOI] [PubMed] [Google Scholar]
  100. Duarte JV, Pereira JM, Quendera B, Raimundo M, Moreno C, Gomes L, Carrilho F, Castelo-Branco M. Early disrupted neurovascular coupling and changed event level hemodynamic response function in type 2 diabetes: an fMRI study. J Cereb Blood Flow Metab. 2015;35:1671–1680. doi: 10.1038/jcbfm.2015.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Dustin ML. Complement Receptors in Myeloid Cell Adhesion and Phagocytosis. Microbiol Spectr. 2016;4 doi: 10.1128/microbiolspec.MCHD-0034-2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Duvanel CB, Honegger P, Pershadsingh H, Feinstein D, Matthieu JM. Inhibition of glial cell proinflammatory activities by peroxisome proliferator-activated receptor gamma agonist confers partial protection during antimyelin oligodendrocyte glycoprotein demyelination in vitro. J Neurosci Res. 2003;71:246–255. doi: 10.1002/jnr.10471. [DOI] [PubMed] [Google Scholar]
  103. Elbrecht A, Chen Y, Cullinan CA, Hayes N, Leibowitz M, Moller DE, Berger J. Molecular cloning, expression and characterization of human peroxisome proliferator activated receptors gamma 1 and gamma 2. Biochem Biophys Res Commun. 1996;224:431–437. doi: 10.1006/bbrc.1996.1044. [DOI] [PubMed] [Google Scholar]
  104. Elder CP, Apple DF, Bickel CS, Meyer RA, Dudley GA. Intramuscular fat and glucose tolerance after spinal cord injury–a cross-sectional study. Spinal Cord. 2004;42:711–716. doi: 10.1038/sj.sc.3101652. [DOI] [PubMed] [Google Scholar]
  105. Elmelund M, Klarskov N, Bagi P, Oturai PS, Biering-Sorensen F. Renal deterioration after spinal cord injury is associated with length of detrusor contractions during cystometry-A study with a median of 41 years follow-up. Neurourol Urodyn. 2017 doi: 10.1002/nau.23163. [DOI] [PubMed] [Google Scholar]
  106. Elting JW, de Jager AE, Teelken AW, Schaaf MJ, Maurits NM, van der Naalt J, Sibinga CT, Sulter GA, De Keyser J. Comparison of serum S-100 protein levels following stroke and traumatic brain injury. J Neurol Sci. 2000;181:104–110. doi: 10.1016/s0022-510x(00)00442-1. [DOI] [PubMed] [Google Scholar]
  107. Erdmann E, Harding S, Lam H, Perez A. Ten-year observational follow-up of PROactive: a randomized cardiovascular outcomes trial evaluating pioglitazone in type 2 diabetes. Diabetes Obes Metab. 2016;18:266–273. doi: 10.1111/dom.12608. [DOI] [PubMed] [Google Scholar]
  108. Erdmann E, Song E, Spanheimer R, van Troostenburg de Bruyn AR, Perez A. Observational follow-up of the PROactive study: a 6-year update. Diabetes Obes Metab. 2014;16:63–74. doi: 10.1111/dom.12180. [DOI] [PubMed] [Google Scholar]
  109. Erdmann E, Spanheimer R, Charbonnel B, Investigators, P.R.S. Pioglitazone and the risk of cardiovascular events in patients with Type 2 diabetes receiving concomitant treatment with nitrates, renin-angiotensin system blockers, or insulin: results from the PROactive study (PROactive 20) J Diabetes. 2010;2:212–220. doi: 10.1111/j.1753-0407.2010.00082.x. [DOI] [PubMed] [Google Scholar]
  110. Ericsson H, Hamren B, Bergstrand S, Elebring M, Fryklund L, Heijer M, Ohman KP. Pharmacokinetics and metabolism of tesaglitazar, a novel dual-acting peroxisome proliferator-activated receptor alpha/gamma agonist, after a single oral and intravenous dose in humans. Drug Metab Dispos. 2004;32:923–929. [PubMed] [Google Scholar]
  111. Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4:1313–1317. doi: 10.1038/3305. [DOI] [PubMed] [Google Scholar]
  112. Ezekowitz RA, Sastry K, Bailly P, Warner A. Molecular characterization of the human macrophage mannose receptor: demonstration of multiple carbohydrate recognition-like domains and phagocytosis of yeasts in Cos-1 cells. J Exp Med. 1990;172:1785–1794. doi: 10.1084/jem.172.6.1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Fadok VA, Warner ML, Bratton DL, Henson PM. CD36 is required for phagocytosis of apoptotic cells by human macrophages that use either a phosphatidylserine receptor or the vitronectin receptor (alpha v beta 3) J Immunol. 1998;161:6250–6257. [PubMed] [Google Scholar]
  114. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, Vidal-Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J. The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem. 1997;272:18779–18789. doi: 10.1074/jbc.272.30.18779. [DOI] [PubMed] [Google Scholar]
  115. Fancy SP, Zhao C, Franklin RJ. Increased expression of Nkx2.2 and Olig2 identifies reactive oligodendrocyte progenitor cells responding to demyelination in the adult CNS. Mol Cell Neurosci. 2004;27:247–254. doi: 10.1016/j.mcn.2004.06.015. [DOI] [PubMed] [Google Scholar]
  116. Fang H, Chen J, Lin S, Wang P, Wang Y, Xiong X, Yang Q. CD36-mediated hematoma absorption following intracerebral hemorrhage: negative regulation by TLR4 signaling. J Immunol. 2014;192:5984–5992. doi: 10.4049/jimmunol.1400054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Faveeuw C, Fougeray S, Angeli V, Fontaine J, Chinetti G, Gosset P, Delerive P, Maliszewski C, Capron M, Staels B, Moser M, Trottein F. Peroxisome proliferator-activated receptor gamma activators inhibit interleukin-12 production in murine dendritic cells. FEBS Lett. 2000;486:261–266. doi: 10.1016/s0014-5793(00)02319-x. [DOI] [PubMed] [Google Scholar]
  118. FDA. FDA Drug Safety Communication: FDA requires removal of some prescribing and dispensing restrictions for rosiglitazone-containing diabetes medicines 2013 [Google Scholar]
  119. FDA. FDA Drug Safety Communication: FDA eliminates the Risk Evaluation and Mitigation Strategy (REMS) for rosiglitazone-containing diabetes medicines 2015 [Google Scholar]
  120. Feddersen RM, Van Ness BG. Single copy gene detection requiring minimal cell numbers. Biotechniques. 1989;7:44–49. [PubMed] [Google Scholar]
  121. Feinstein DL, Galea E, Gavrilyuk V, Brosnan CF, Whitacre CC, Dumitrescu-Ozimek L, Landreth GE, Pershadsingh HA, Weinberg G, Heneka MT. Peroxisome proliferator-activated receptor-gamma agonists prevent experimental autoimmune encephalomyelitis. Ann Neurol. 2002;51:694–702. doi: 10.1002/ana.10206. [DOI] [PubMed] [Google Scholar]
  122. Ferguson LB, Most D, Blednov YA, Harris RA. PPAR agonists regulate brain gene expression: relationship to their effects on ethanol consumption. Neuropharmacology. 2014;86:397–407. doi: 10.1016/j.neuropharm.2014.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Finch ER, Tukaramrao DB, Goodfield LL, Quickel MD, Paulson RF, Prabhu KS. Activation of PPARgamma by endogenous prostaglandin J2 mediates the antileukemic effect of selenium in murine leukemia. Blood. 2017;129:1802–1810. doi: 10.1182/blood-2016-08-736405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Floyd ZE, Wang ZQ, Kilroy G, Cefalu WT. Modulation of peroxisome proliferator-activated receptor gamma stability and transcriptional activity in adipocytes by resveratrol. Metabolism. 2008;57:S32–38. doi: 10.1016/j.metabol.2008.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Flygt J, Djupsjo A, Lenne F, Marklund N. Myelin loss and oligodendrocyte pathology in white matter tracts following traumatic brain injury in the rat. Eur J Neurosci. 2013;38:2153–2165. doi: 10.1111/ejn.12179. [DOI] [PubMed] [Google Scholar]
  126. Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell. 1995;83:803–812. doi: 10.1016/0092-8674(95)90193-0. [DOI] [PubMed] [Google Scholar]
  127. Frankel HL, Coll JR, Charlifue SW, Whiteneck GG, Gardner BP, Jamous MA, Krishnan KR, Nuseibeh I, Savic G, Sett P. Long-term survival in spinal cord injury: a fifty year investigation. Spinal Cord. 1998;36:266–274. doi: 10.1038/sj.sc.3100638. [DOI] [PubMed] [Google Scholar]
  128. Franklin RJ, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci. 2008;9:839–855. doi: 10.1038/nrn2480. [DOI] [PubMed] [Google Scholar]
  129. Freeman SA, Grinstein S. Phagocytosis: receptors, signal integration, and the cytoskeleton. Immunol Rev. 2014;262:193–215. doi: 10.1111/imr.12212. [DOI] [PubMed] [Google Scholar]
  130. Funfschilling U, Supplie LM, Mahad D, Boretius S, Saab AS, Edgar J, Brinkmann BG, Kassmann CM, Tzvetanova ID, Mobius W, Diaz F, Meijer D, Suter U, Hamprecht B, Sereda MW, Moraes CT, Frahm J, Goebbels S, Nave KA. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature. 2012;485:517–521. doi: 10.1038/nature11007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Gaetani P, Marzatico F, Rodriguez y Baena R, Pacchiarini L, Vigano T, Grignani G, Crivellari MT, Benzi G. Arachidonic acid metabolism and pathophysiologic aspects of subarachnoid hemorrhage in rats. Stroke. 1990;21:328–332. doi: 10.1161/01.str.21.2.328. [DOI] [PubMed] [Google Scholar]
  132. Galeffi F, Degan S, Britz G, Turner DA. Dysregulation of oxygen hemodynamic responses to synaptic train stimulation in a rat hippocampal model of subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2016;36:696–701. doi: 10.1177/0271678X15624699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Gan Y, Liu Q, Wu W, Yin JX, Bai XF, Shen R, Wang Y, Chen J, La Cava A, Poursine-Laurent J, Yokoyama W, Shi FD. Ischemic neurons recruit natural killer cells that accelerate brain infarction. Proc Natl Acad Sci U S A. 2014;111:2704–2709. doi: 10.1073/pnas.1315943111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Garcia-Bueno B, Caso JR, Perez-Nievas BG, Lorenzo P, Leza JC. Effects of peroxisome proliferator-activated receptor gamma agonists on brain glucose and glutamate transporters after stress in rats. Neuropsychopharmacology. 2007;32:1251–1260. doi: 10.1038/sj.npp.1301252. [DOI] [PubMed] [Google Scholar]
  135. Gelderblom M, Leypoldt F, Steinbach K, Behrens D, Choe CU, Siler DA, Arumugam TV, Orthey E, Gerloff C, Tolosa E, Magnus T. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857. doi: 10.1161/STROKEAHA.108.534503. [DOI] [PubMed] [Google Scholar]
  136. Gennarelli TA. Mechanisms of brain injury. J Emerg Med. 1993;11(Suppl 1):5–11. [PubMed] [Google Scholar]
  137. Genovese T, Esposito E, Mazzon E, Di Paola R, Muia C, Meli R, Bramanti P, Cuzzocrea S. Effect of cyclopentanone prostaglandin 15-deoxy-delta12, 14PGJ2 on early functional recovery from experimental spinal cord injury. Shock. 2008;30:142–152. doi: 10.1097/SHK.0b013e31815dd381. [DOI] [PubMed] [Google Scholar]
  138. Gensert JM, Goldman JE. Endogenous progenitors remyelinate demyelinated axons in the adult CNS. Neuron. 1997;19:197–203. doi: 10.1016/s0896-6273(00)80359-1. [DOI] [PubMed] [Google Scholar]
  139. Ghajar J. Traumatic brain injury. Lancet. 2000;356:923–929. doi: 10.1016/S0140-6736(00)02689-1. [DOI] [PubMed] [Google Scholar]
  140. Ghirnikar RS, Lee YL, Eng LF. Inflammation in traumatic brain injury: role of cytokines and chemokines. Neurochem Res. 1998;23:329–340. doi: 10.1023/a:1022453332560. [DOI] [PubMed] [Google Scholar]
  141. Gillespie W, Tyagi N, Tyagi SC. Role of PPARgamma, a nuclear hormone receptor in neuroprotection. Indian J Biochem Biophys. 2011;48:73–81. [PubMed] [Google Scholar]
  142. Girnun GD, Domann FE, Moore SA, Robbins ME. Identification of a functional peroxisome proliferator-activated receptor response element in the rat catalase promoter. Mol Endocrinol. 2002;16:2793–2801. doi: 10.1210/me.2002-0020. [DOI] [PubMed] [Google Scholar]
  143. Glass CK, Ogawa S. Combinatorial roles of nuclear receptors in inflammation and immunity. Nat Rev Immunol. 2006;6:44–55. doi: 10.1038/nri1748. [DOI] [PubMed] [Google Scholar]
  144. Gold M, Alderton C, Zvartau-Hind M, Egginton S, Saunders AM, Irizarry M, Craft S, Landreth G, Linnamagi U, Sawchak S. Rosiglitazone monotherapy in mild-to-moderate Alzheimer’s disease: results from a randomized, double-blind, placebo-controlled phase III study. Dement Geriatr Cogn Disord. 2010;30:131–146. doi: 10.1159/000318845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Gong C, Hoff JT, Keep RF. Acute inflammatory reaction following experimental intracerebral hemorrhage in rat. Brain Res. 2000;871:57–65. doi: 10.1016/s0006-8993(00)02427-6. [DOI] [PubMed] [Google Scholar]
  146. Gosset P, Charbonnier AS, Delerive P, Fontaine J, Staels B, Pestel J, Tonnel AB, Trottein F. Peroxisome proliferator-activated receptor gamma activators affect the maturation of human monocyte-derived dendritic cells. Eur J Immunol. 2001;31:2857–2865. doi: 10.1002/1521-4141(2001010)31:10<2857::aid-immu2857>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  147. Gough PJ, Gomez IG, Wille PT, Raines EW. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest. 2006;116:59–69. doi: 10.1172/JCI25074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Goverman J. Autoimmune T cell responses in the central nervous system. Nat Rev Immunol. 2009;9:393–407. doi: 10.1038/nri2550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Graham DJ, Ouellet-Hellstrom R, MaCurdy TE, Ali F, Sholley C, Worrall C, Kelman JA. Risk of acute myocardial infarction, stroke, heart failure, and death in elderly Medicare patients treated with rosiglitazone or pioglitazone. JAMA. 2010;304:411–418. doi: 10.1001/jama.2010.920. [DOI] [PubMed] [Google Scholar]
  150. Grau AJ, Urbanek C, Palm F. Common infections and the risk of stroke. Nat Rev Neurol. 2010;6:681–694. doi: 10.1038/nrneurol.2010.163. [DOI] [PubMed] [Google Scholar]
  151. Greene ME, Blumberg B, McBride OW, Yi HF, Kronquist K, Kwan K, Hsieh L, Greene G, Nimer SD. Isolation of the human peroxisome proliferator activated receptor gamma cDNA: expression in hematopoietic cells and chromosomal mapping. Gene Expr. 1995;4:281–299. [PMC free article] [PubMed] [Google Scholar]
  152. Greenhalgh AD, David S. Differences in the phagocytic response of microglia and peripheral macrophages after spinal cord injury and its effects on cell death. J Neurosci. 2014;34:6316–6322. doi: 10.1523/JNEUROSCI.4912-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Griggs RB, Donahue RR, Morgenweck J, Grace PM, Sutton A, Watkins LR, Taylor BK. Pioglitazone rapidly reduces neuropathic pain through astrocyte and nongenomic PPARgamma mechanisms. Pain. 2015;156:469–482. doi: 10.1097/01.j.pain.0000460333.79127.be. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Grommes C, Karlo JC, Caprariello A, Blankenship D, Dechant A, Landreth GE. The PPARgamma agonist pioglitazone crosses the blood-brain barrier and reduces tumor growth in a human xenograft model. Cancer Chemother Pharmacol. 2013;71:929–936. doi: 10.1007/s00280-013-2084-2. [DOI] [PubMed] [Google Scholar]
  155. Groves AK, Barnett SC, Franklin RJ, Crang AJ, Mayer M, Blakemore WF, Noble M. Repair of demyelinated lesions by transplantation of purified O-2A progenitor cells. Nature. 1993;362:453–455. doi: 10.1038/362453a0. [DOI] [PubMed] [Google Scholar]
  156. Grygiel-Gorniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications–a review. Nutr J. 2014;13:17. doi: 10.1186/1475-2891-13-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Guan LM, Zhao J, Scandalios JG. Cis-elements and trans-factors that regulate expression of the maize Cat1 antioxidant gene in response to ABA and osmotic stress: H2O2 is the likely intermediary signaling molecule for the response. Plant J. 2000;22:87–95. doi: 10.1046/j.1365-313x.2000.00723.x. [DOI] [PubMed] [Google Scholar]
  158. Guan Y, Zhang Y, Davis L, Breyer MD. Expression of peroxisome proliferator-activated receptors in urinary tract of rabbits and humans. Am J Physiol. 1997;273:F1013–1022. doi: 10.1152/ajprenal.1997.273.6.F1013. [DOI] [PubMed] [Google Scholar]
  159. Habib ZA, Tzogias L, Havstad SL, Wells K, Divine G, Lanfear DE, Tang J, Krajenta R, Pladevall M, Williams LK. Relationship between thiazolidinedione use and cardiovascular outcomes and all-cause mortality among patients with diabetes: a time-updated propensity analysis. Pharmacoepidemiol Drug Saf. 2009;18:437–447. doi: 10.1002/pds.1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol. 1996;53:309–315. doi: 10.1001/archneur.1996.00550040037012. [DOI] [PubMed] [Google Scholar]
  161. Halabi CM, Beyer AM, de Lange WJ, Keen HL, Baumbach GL, Faraci FM, Sigmund CD. Interference with PPAR gamma function in smooth muscle causes vascular dysfunction and hypertension. Cell Metab. 2008;7:215–226. doi: 10.1016/j.cmet.2007.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Hamblin M, Chang L, Fan Y, Zhang J, Chen YE. PPARs and the cardiovascular system. Antioxid Redox Signal. 2009;11:1415–1452. doi: 10.1089/ars.2008.2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Hamming AM, Wermer MJ, Umesh Rudrapatna S, Lanier C, van Os HJ, van den Bergh WM, Ferrari MD, van der Toorn A, van den Maagdenberg AM, Stowe AM, Dijkhuizen RM. Spreading depolarizations increase delayed brain injury in a rat model of subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2016;36:1224–1231. doi: 10.1177/0271678X15619189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Han L, Cai W, Mao L, Liu J, Li P, Leak RK, Xu Y, Hu X, Chen J. Rosiglitazone Promotes White Matter Integrity and Long-Term Functional Recovery After Focal Cerebral Ischemia. Stroke. 2015;46:2628–2636. doi: 10.1161/STROKEAHA.115.010091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Harrison BM, Gledhill RF, McDonald WJ. Remyelination after transient compression of the spinal cord. Proc Aust Assoc Neurol. 1975;12:117–122. [PubMed] [Google Scholar]
  166. Hawkins CP, Mackenzie F, Tofts P, du Boulay EP, McDonald WI. Patterns of blood-brain barrier breakdown in inflammatory demyelination. Brain. 1991;114(Pt 2):801–810. doi: 10.1093/brain/114.2.801. [DOI] [PubMed] [Google Scholar]
  167. He F, Doucet JA, Stephens JM. Caspase-mediated degradation of PPARgamma proteins in adipocytes. Obesity (Silver Spring) 2008;16:1735–1741. doi: 10.1038/oby.2008.269. [DOI] [PubMed] [Google Scholar]
  168. Heinaniemi M, Uski JO, Degenhardt T, Carlberg C. Meta-analysis of primary target genes of peroxisome proliferator- activated receptors. Genome Biol. 2007;8:R147. doi: 10.1186/gb-2007-8-7-r147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Heneka MT, Feinstein DL, Galea E, Gleichmann M, Wullner U, Klockgether T. Peroxisome proliferator-activated receptor gamma agonists protect cerebellar granule cells from cytokine-induced apoptotic cell death by inhibition of inducible nitric oxide synthase. J Neuroimmunol. 1999;100:156–168. doi: 10.1016/s0165-5728(99)00192-7. [DOI] [PubMed] [Google Scholar]
  170. Henson PM, Hume DA. Apoptotic cell removal in development and tissue homeostasis. Trends Immunol. 2006;27:244–250. doi: 10.1016/j.it.2006.03.005. [DOI] [PubMed] [Google Scholar]
  171. Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358–372. doi: 10.1038/nrn3880. [DOI] [PubMed] [Google Scholar]
  172. Hesp ZC, Goldstein EZ, Miranda CJ, Kaspar BK, McTigue DM. Chronic oligodendrogenesis and remyelination after spinal cord injury in mice and rats. J Neurosci. 2015;35:1274–1290. doi: 10.1523/JNEUROSCI.2568-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Hickenbottom SL, Grotta JC, Strong R, Denner LA, Aronowski J. Nuclear factor-kappaB and cell death after experimental intracerebral hemorrhage in rats. Stroke. 1999;30:2472–2477. doi: 10.1161/01.str.30.11.2472. discussion 2477-2478. [DOI] [PubMed] [Google Scholar]
  174. Hills NK, Johnston SC, Sidney S, Zielinski BA, Fullerton HJ. Recent trauma and acute infection as risk factors for childhood arterial ischemic stroke. Ann Neurol. 2012;72:850–858. doi: 10.1002/ana.23688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Hills NK, Sidney S, Fullerton HJ. Timing and number of minor infections as risk factors for childhood arterial ischemic stroke. Neurology. 2014;83:890–897. doi: 10.1212/WNL.0000000000000752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Hind WH, Tufarelli C, Neophytou M, Anderson SI, England TJ, O’Sullivan SE. Endocannabinoids modulate human blood-brain barrier permeability in vitro. Br J Pharmacol. 2015;172:3015–3027. doi: 10.1111/bph.13106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ, Team, R.S. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet. 2009;373:2125–2135. doi: 10.1016/S0140-6736(09)60953-3. [DOI] [PubMed] [Google Scholar]
  178. Hovelmeyer N, Hao Z, Kranidioti K, Kassiotis G, Buch T, Frommer F, von Hoch L, Kramer D, Minichiello L, Kollias G, Lassmann H, Waisman A. Apoptosis of oligodendrocytes via Fas and TNF-R1 is a key event in the induction of experimental autoimmune encephalomyelitis. J Immunol. 2005;175:5875–5884. doi: 10.4049/jimmunol.175.9.5875. [DOI] [PubMed] [Google Scholar]
  179. Howard G, Evans GW, Pearce K, Howard VJ, Bell RA, Mayer EJ, Burke GL. Is the stroke belt disappearing? An analysis of racial, temporal, and age effects. Stroke. 1995;26:1153–1158. doi: 10.1161/01.str.26.7.1153. [DOI] [PubMed] [Google Scholar]
  180. Hsiao FY, Huang WF, Wen YW, Chen PF, Kuo KN, Tsai YW. Thiazolidinediones and cardiovascular events in patients with type 2 diabetes mellitus: a retrospective cohort study of over 473,000 patients using the National Health Insurance database in Taiwan. Drug Saf. 2009;32:675–690. doi: 10.2165/00002018-200932080-00006. [DOI] [PubMed] [Google Scholar]
  181. Hu E, Kim JB, Sarraf P, Spiegelman BM. Inhibition of adipogenesis through MAP kinase-mediated phosphorylation of PPARgamma. Science. 1996;274:2100–2103. doi: 10.1126/science.274.5295.2100. [DOI] [PubMed] [Google Scholar]
  182. Hu X, Leak RK, Shi Y, Suenaga J, Gao Y, Zheng P, Chen J. Microglial and macrophage polarization-new prospects for brain repair. Nat Rev Neurol. 2015;11:56–64. doi: 10.1038/nrneurol.2014.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, Gao Y, Chen J. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke. 2012;43:3063–3070. doi: 10.1161/STROKEAHA.112.659656. [DOI] [PubMed] [Google Scholar]
  184. Huria T, Beeraka NM, Al-Ghamdi B, Fern R. Premyelinated central axons express neurotoxic NMDA receptors: relevance to early developing white-matter injury. J Cereb Blood Flow Metab. 2015;35:543–553. doi: 10.1038/jcbfm.2014.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Husain J, Juurlink BH. Oligodendroglial precursor cell susceptibility to hypoxia is related to poor ability to cope with reactive oxygen species. Brain Res. 1995;698:86–94. doi: 10.1016/0006-8993(95)00832-b. [DOI] [PubMed] [Google Scholar]
  186. Huxley RR, Barzi F, Lam TH, Czernichow S, Fang X, Welborn T, Shaw J, Ueshima H, Zimmet P, Jee SH, Patel JV, Caterson I, Perkovic V, Woodward M, Asia Pacific Cohort Studies, C. the Obesity in Asia, C Isolated low levels of high-density lipoprotein cholesterol are associated with an increased risk of coronary heart disease: an individual participant data meta-analysis of 23 studies in the Asia-Pacific region. Circulation. 2011;124:2056–2064. doi: 10.1161/CIRCULATIONAHA.111.028373. [DOI] [PubMed] [Google Scholar]
  187. Iankova I, Petersen RK, Annicotte JS, Chavey C, Hansen JB, Kratchmarova I, Sarruf D, Benkirane M, Kristiansen K, Fajas L. Peroxisome proliferator-activated receptor gamma recruits the positive transcription elongation factor b complex to activate transcription and promote adipogenesis. Mol Endocrinol. 2006;20:1494–1505. doi: 10.1210/me.2005-0222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Ikeda Y, Sugawara A, Taniyama Y, Uruno A, Igarashi K, Arima S, Ito S, Takeuchi K. Suppression of rat thromboxane synthase gene transcription by peroxisome proliferator-activated receptor gamma in macrophages via an interaction with NRF2. J Biol Chem. 2000;275:33142–33150. doi: 10.1074/jbc.M002319200. [DOI] [PubMed] [Google Scholar]
  189. Imamoto E, Yoshida N, Uchiyama K, Kuroda M, Kokura S, Ichikawa H, Naito Y, Tanigawa T, Yoshikawa T. Inhibitory effect of pioglitazone on expression of adhesion molecules on neutrophils and endothelial cells. Biofactors. 2004;20:37–47. doi: 10.1002/biof.5520200104. [DOI] [PubMed] [Google Scholar]
  190. Inzucchi SE, Furie KL. The IRIS (Insulin Resistance Intervention after Stroke) trial: A new perspective on pioglitazone. J Diabetes. 2016;8:607–609. doi: 10.1111/1753-0407.12399. [DOI] [PubMed] [Google Scholar]
  191. Irvine KA, Blakemore WF. Remyelination protects axons from demyelination-associated axon degeneration. Brain. 2008;131:1464–1477. doi: 10.1093/brain/awn080. [DOI] [PubMed] [Google Scholar]
  192. Ishii T, Itoh K, Ruiz E, Leake DS, Unoki H, Yamamoto M, Mann GE. Role of Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages: activation by oxidatively modified LDL and 4-hydroxynonenal. Circ Res. 2004;94:609–616. doi: 10.1161/01.RES.0000119171.44657.45. [DOI] [PubMed] [Google Scholar]
  193. Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 1990;347:645–650. doi: 10.1038/347645a0. [DOI] [PubMed] [Google Scholar]
  194. Iwai S, Maeda T, Kiguchi N, Kobayashi Y, Fukazawa Y, Ozaki M, Kishioka S. Pioglitazone attenuates tactile allodynia and microglial activation in mice with peripheral nerve injury. Drug Discov Ther. 2008;2:353–356. [PubMed] [Google Scholar]
  195. Izawa Y, Takahashi S, Suzuki N. Pioglitazone enhances pyruvate and lactate oxidation in cultured neurons but not in cultured astroglia. Brain Res. 2009;1305:64–73. doi: 10.1016/j.brainres.2009.09.098. [DOI] [PubMed] [Google Scholar]
  196. Jackson SM, Parhami F, Xi XP, Berliner JA, Hsueh WA, Law RE, Demer LL. Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction. Arterioscler Thromb Vasc Biol. 1999;19:2094–2104. doi: 10.1161/01.atv.19.9.2094. [DOI] [PubMed] [Google Scholar]
  197. Jana M, Mondal S, Gonzalez FJ, Pahan K. Gemfibrozil, a lipid-lowering drug, increases myelin genes in human oligodendrocytes via peroxisome proliferator-activated receptor-beta. J Biol Chem. 2012;287:34134–34148. doi: 10.1074/jbc.M112.398552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Janani C, Ranjitha Kumari BD. PPAR gamma gene–a review. Diabetes Metab Syndr. 2015;9:46–50. doi: 10.1016/j.dsx.2014.09.015. [DOI] [PubMed] [Google Scholar]
  199. Jeffery ND, Blakemore WF. Locomotor deficits induced by experimental spinal cord demyelination are abolished by spontaneous remyelination. Brain. 1997;120(Pt 1):27–37. doi: 10.1093/brain/120.1.27. [DOI] [PubMed] [Google Scholar]
  200. Jennewein C, Kuhn AM, Schmidt MV, Meilladec-Jullig V, von Knethen A, Gonzalez FJ, Brune B. Sumoylation of peroxisome proliferator-activated receptor gamma by apoptotic cells prevents lipopolysaccharide-induced NCoR removal from kappaB binding sites mediating transrepression of proinflammatory cytokines. J Immunol. 2008;181:5646–5652. doi: 10.4049/jimmunol.181.8.5646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Jeremitsky E, Omert LA, Dunham CM, Wilberger J, Rodriguez A. The impact of hyperglycemia on patients with severe brain injury. J Trauma. 2005;58:47–50. doi: 10.1097/01.ta.0000135158.42242.b1. [DOI] [PubMed] [Google Scholar]
  202. Jia HB, Wang XM, Qiu LL, Liu XY, Shen JC, Ji Q, Yang JJ. Spinal neuroimmune activation inhibited by repeated administration of pioglitazone in rats after L5 spinal nerve transection. Neurosci Lett. 2013;543:130–135. doi: 10.1016/j.neulet.2013.03.046. [DOI] [PubMed] [Google Scholar]
  203. Jiang X, Pu H, Hu X, Wei Z, Hong D, Zhang W, Gao Y, Chen J, Shi Y. A Post-stroke Therapeutic Regimen with Omega-3 Polyunsaturated Fatty Acids that Promotes White Matter Integrity and Beneficial Microglial Responses after Cerebral Ischemia. Transl Stroke Res. 2016;7:548–561. doi: 10.1007/s12975-016-0502-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Jin K, Wang X, Xie L, Mao XO, Greenberg DA. Transgenic ablation of doublecortin-expressing cells suppresses adult neurogenesis and worsens stroke outcome in mice. Proc Natl Acad Sci U S A. 2010a;107:7993–7998. doi: 10.1073/pnas.1000154107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010b;87:779–789. doi: 10.1189/jlb.1109766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Jin WN, Shi SX, Li Z, Li M, Wood K, Gonzales RJ, Liu Q. Depletion of microglia exacerbates postischemic inflammation and brain injury. J Cereb Blood Flow Metab. 2017;37:2224–2236. doi: 10.1177/0271678X17694185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Frisen J. Identification of a neural stem cell in the adult mammalian central nervous system. Cell. 1999;96:25–34. doi: 10.1016/s0092-8674(00)80956-3. [DOI] [PubMed] [Google Scholar]
  208. Johnstone JT, Morton PD, Jayakumar AR, Johnstone AL, Gao H, Bracchi-Ricard V, Pearse DD, Norenberg MD, Bethea JR. Inhibition of NADPH oxidase activation in oligodendrocytes reduces cytotoxicity following trauma. PLoS One. 2013;8:e80975. doi: 10.1371/journal.pone.0080975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Jordan JE, Zhao ZQ, Vinten-Johansen J. The role of neutrophils in myocardial ischemia-reperfusion injury. Cardiovasc Res. 1999;43:860–878. doi: 10.1016/s0008-6363(99)00187-x. [DOI] [PubMed] [Google Scholar]
  210. Joseph MJ, Caliaperumal J, Schlichter LC. After Intracerebral Hemorrhage, Oligodendrocyte Precursors Proliferate and Differentiate Inside White-Matter Tracts in the Rat Striatum. Transl Stroke Res. 2016;7:192–208. doi: 10.1007/s12975-015-0445-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Jurkunas UV, Bitar MS, Funaki T, Azizi B. Evidence of oxidative stress in the pathogenesis of fuchs endothelial corneal dystrophy. Am J Pathol. 2010;177:2278–2289. doi: 10.2353/ajpath.2010.100279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Juurlink BH. Response of glial cells to ischemia: roles of reactive oxygen species and glutathione. Neurosci Biobehav Rev. 1997;21:151–166. doi: 10.1016/s0149-7634(96)00005-x. [DOI] [PubMed] [Google Scholar]
  213. Kanakasabai S, Pestereva E, Chearwae W, Gupta SK, Ansari S, Bright JJ. PPARgamma agonists promote oligodendrocyte differentiation of neural stem cells by modulating stemness and differentiation genes. PLoS One. 2012;7:e50500. doi: 10.1371/journal.pone.0050500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Kane CJ, Phelan KD, Han L, Smith RR, Xie J, Douglas JC, Drew PD. Protection of neurons and microglia against ethanol in a mouse model of fetal alcohol spectrum disorders by peroxisome proliferator-activated receptor-gamma agonists. Brain Behav Immun. 2011;25(Suppl 1):S137–145. doi: 10.1016/j.bbi.2011.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Kapadia R, Yi JH, Vemuganti R. Mechanisms of anti-inflammatory and neuroprotective actions of PPAR-gamma agonists. Front Biosci. 2008;13:1813–1826. doi: 10.2741/2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Kaplan JM, Cook JA, Hake PW, O’Connor M, Burroughs TJ, Zingarelli B. 15-Deoxy-delta(12,14)-prostaglandin J(2) (15D-PGJ(2)), a peroxisome proliferator activated receptor gamma ligand, reduces tissue leukosequestration and mortality in endotoxic shock. Shock. 2005;24:59–65. doi: 10.1097/01.shk.0000167108.88376.f2. [DOI] [PubMed] [Google Scholar]
  217. Katzan I. Can add-on pioglitazone prevent recurrent stroke in patients with type 2 diabetes? Nat Clin Pract Neurol. 2007;3:486–487. doi: 10.1038/ncpneuro0562. [DOI] [PubMed] [Google Scholar]
  218. Kaundal RK, Sharma SS. Peroxisome proliferator-activated receptor gamma agonists as neuroprotective agents. Drug News Perspect. 2010;23:241–256. doi: 10.1358/dnp.2010.23.4.1437710. [DOI] [PubMed] [Google Scholar]
  219. Kawabori M, Yenari MA. Inflammatory responses in brain ischemia. Curr Med Chem. 2015;22:1258–1277. doi: 10.2174/0929867322666150209154036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr, Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O’Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR, Investigators, I.T. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med. 2016;374:1321–1331. doi: 10.1056/NEJMoa1506930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Khandoudi N, Delerive P, Berrebi-Bertrand I, Buckingham RE, Staels B, Bril A. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma, inhibits the Jun NH(2)-terminal kinase/activating protein 1 pathway and protects the heart from ischemia/reperfusion injury. Diabetes. 2002;51:1507–1514. doi: 10.2337/diabetes.51.5.1507. [DOI] [PubMed] [Google Scholar]
  222. Kheir WA, Gevrey JC, Yamaguchi H, Isaac B, Cox D. A WAVE2-Abi1 complex mediates CSF-1-induced F-actin-rich membrane protrusions and migration in macrophages. J Cell Sci. 2005;118:5369–5379. doi: 10.1242/jcs.02638. [DOI] [PubMed] [Google Scholar]
  223. Kielian T, Syed MM, Liu S, Phulwani NK, Phillips N, Wagoner G, Drew PD, Esen N. The synthetic peroxisome proliferator-activated receptor-gamma agonist ciglitazone attenuates neuroinflammation and accelerates encapsulation in b0Oacterial brain abscesses. J Immunol. 2008;180:5004–5016. doi: 10.4049/jimmunol.180.7.5004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Kim TH, Kim MY, Jo SH, Park JM, Ahn YH. Modulation of the transcriptional activity of peroxisome proliferator-activated receptor gamma by protein-protein interactions and post-translational modifications. Yonsei Med J. 2013;54:545–559. doi: 10.3349/ymj.2013.54.3.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Kivisakk P, Imitola J, Rasmussen S, Elyaman W, Zhu B, Ransohoff RM, Khoury SJ. Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann Neurol. 2009;65:457–469. doi: 10.1002/ana.21379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Klopfleisch S, Merkler D, Schmitz M, Kloppner S, Schedensack M, Jeserich G, Althaus HH, Bruck W. Negative impact of statins on oligodendrocytes and myelin formation in vitro and in vivo. J Neurosci. 2008;28:13609–13614. doi: 10.1523/JNEUROSCI.2765-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S, Alferink J, Nowak N, Beyer M, Mayer G, Langhans B, Klockgether T, Waisman A, Eberl G, Schultze J, Famulok M, Kolanus W, Glass C, Kurts C, Knolle PA. The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med. 2009;206:2079–2089. doi: 10.1084/jem.20082771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Koenigsknecht-Talboo J, Landreth GE. Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci. 2005;25:8240–8249. doi: 10.1523/JNEUROSCI.1808-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi BV, Jacobson KA, Kohsaka S, Inoue K. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446:1091–1095. doi: 10.1038/nature05704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Kokoeva MV, Yin H, Flier JS. Neurogenesis in the hypothalamus of adult mice: potential role in energy balance. Science. 2005;310:679–683. doi: 10.1126/science.1115360. [DOI] [PubMed] [Google Scholar]
  231. Krueger M, Bechmann I, Immig K, Reichenbach A, Hartig W, Michalski D. Blood-brain barrier breakdown involves four distinct stages of vascular damage in various models of experimental focal cerebral ischemia. J Cereb Blood Flow Metab. 2015;35:292–303. doi: 10.1038/jcbfm.2014.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Kumari R, Willing LB, Patel SD, Krady JK, Zavadoski WJ, Gibbs EM, Vannucci SJ, Simpson IA. The PPAR-654 gamma agonist, darglitazone, restores acute inflammatory responses to cerebral hypoxia-ischemia in the diabetic ob/ob mouse. J Cereb Blood Flow Metab. 2010;30:352–360. doi: 10.1038/jcbfm.2009.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–188. doi: 10.1016/j.pneurobio.2013.11.006. [DOI] [PubMed] [Google Scholar]
  234. Lalloyer F, Staels B. Fibrates, glitazones, and peroxisome proliferator-activated receptors. Arterioscler Thromb Vasc Biol. 2010;30:894–899. doi: 10.1161/ATVBAHA.108.179689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Lan LF, Zheng L, Yang X, Ji XT, Fan YH, Zeng JS. Peroxisome proliferator-activated receptor-gamma agonist pioglitazone ameliorates white matter lesion and cognitive impairment in hypertensive rats. CNS Neurosci Ther. 2015;21:410–416. doi: 10.1111/cns.12374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Lee-Liu D, Edwards-Faret G, Tapia VS, Larrain J. Spinal cord regeneration: lessons for mammals from non-mammalian vertebrates. Genesis. 2013;51:529–544. doi: 10.1002/dvg.22406. [DOI] [PubMed] [Google Scholar]
  237. Lee J, Reding M. Effects of thiazolidinediones on stroke recovery: a case-matched controlled study. Neurochem Res. 2007;32:635–638. doi: 10.1007/s11064-006-9138-3. [DOI] [PubMed] [Google Scholar]
  238. Lee TM, Chou TF. Troglitazone administration limits infarct size by reduced phosphorylation of canine myocardial connexin43 proteins. Am J Physiol Heart Circ Physiol. 2003;285:H1650–1659. doi: 10.1152/ajpheart.00407.2002. [DOI] [PubMed] [Google Scholar]
  239. Lee Y, Morrison BM, Li Y, Lengacher S, Farah MH, Hoffman PN, Liu Y, Tsingalia A, Jin L, Zhang PW, Pellerin L, Magistretti PJ, Rothstein JD. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature. 2012;487:443–448. doi: 10.1038/nature11314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Leesnitzer LM, Parks DJ, Bledsoe RK, Cobb JE, Collins JL, Consler TG, Davis RG, Hull-Ryde EA, Lenhard JM, Patel L, Plunket KD, Shenk JL, Stimmel JB, Therapontos C, Willson TM, Blanchard SG. Functional consequences of cysteine modification in the ligand binding sites of peroxisome proliferator activated receptors by GW9662. Biochemistry. 2002;41:6640–6650. doi: 10.1021/bi0159581. [DOI] [PubMed] [Google Scholar]
  241. Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma) J Biol Chem. 1995;270:12953–12956. doi: 10.1074/jbc.270.22.12953. [DOI] [PubMed] [Google Scholar]
  242. Leisewitz AV, Urrutia CR, Martinez GR, Loyola G, Bronfman M. A PPARs cross-talk concertedly commits C6 glioma cells to oligodendrocytes and induces enzymes involved in myelin synthesis. J Cell Physiol. 2008;217:367–376. doi: 10.1002/jcp.21509. [DOI] [PubMed] [Google Scholar]
  243. Lemay DG, Hwang DH. Genome-wide identification of peroxisome proliferator response elements using integrated computational genomics. J Lipid Res. 2006;47:1583–1587. doi: 10.1194/jlr.M500504-JLR200. [DOI] [PubMed] [Google Scholar]
  244. Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK. Peroxisome proliferator-activated receptor gamma ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2000;106:523–531. doi: 10.1172/JCI10370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Li X, Du J, Xu S, Lin X, Ling Z. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone reduces secondary damage in experimental spinal cord injury. J Int Med Res. 2013;41:153–161. doi: 10.1177/0300060513476601. [DOI] [PubMed] [Google Scholar]
  246. Li X, Melief E, Postupna N, Montine KS, Keene CD, Montine TJ. Prostaglandin E2 receptor subtype 2 regulation of scavenger receptor CD36 modulates microglial Abeta42 phagocytosis. Am J Pathol. 2015;185:230–239. doi: 10.1016/j.ajpath.2014.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Liebetanz D, Merkler D. Effects of commissural de- and remyelination on motor skill behaviour in the cuprizone mouse model of multiple sclerosis. Exp Neurol. 2006;202:217–224. doi: 10.1016/j.expneurol.2006.05.032. [DOI] [PubMed] [Google Scholar]
  248. Liesz A, Dalpke A, Mracsko E, Antoine DJ, Roth S, Zhou W, Yang H, Na SY, Akhisaroglu M, Fleming T, Eigenbrod T, Nawroth PP, Tracey KJ, Veltkamp R. DAMP signaling is a key pathway inducing immune modulation after brain injury. J Neurosci. 2015;35:583–598. doi: 10.1523/JNEUROSCI.2439-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Lin TN, Cheung WM, Wu JS, Chen JJ, Lin H, Chen JJ, Liou JY, Shyue SK, Wu KK. 15d-prostaglandin J2 protects brain from ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol. 2006;26:481–487. doi: 10.1161/01.ATV.0000201933.53964.5b. [DOI] [PubMed] [Google Scholar]
  250. Lin W, Harding HP, Ron D, Popko B. Endoplasmic reticulum stress modulates the response of myelinating oligodendrocytes to the immune cytokine interferon-gamma. J Cell Biol. 2005;169:603–612. doi: 10.1083/jcb.200502086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Lindvall O, Kokaia Z. Neurogenesis following Stroke Affecting the Adult Brain. Cold Spring Harb Perspect Biol. 2015;7 doi: 10.1101/cshperspect.a019034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Liu H, Rose ME, Culver S, Ma X, Dixon CE, Graham SH. Rosiglitazone attenuates inflammation and CA3 neuronal loss following traumatic brain injury in rats. Biochem Biophys Res Commun. 2016;472:648–655. doi: 10.1016/j.bbrc.2016.03.003. [DOI] [PubMed] [Google Scholar]
  253. Liu HR, Tao L, Gao E, Lopez BL, Christopher TA, Willette RN, Ohlstein EH, Yue TL, Ma XL. Anti-apoptotic effects of rosiglitazone in hypercholesterolemic rabbits subjected to myocardial ischemia and reperfusion. Cardiovasc Res. 2004;62:135–144. doi: 10.1016/j.cardiores.2003.12.027. [DOI] [PubMed] [Google Scholar]
  254. Liu J, Wang LN. Peroxisome proliferator-activated receptor gamma agonists for preventing recurrent stroke and other vascular events in patients with stroke or transient ischaemic attack. Cochrane Database Syst Rev. 2015:CD010693. doi: 10.1002/14651858.CD010693.pub3. [DOI] [PubMed] [Google Scholar]
  255. Liu SH, Yang CN, Pan HC, Sung YJ, Liao KK, Chen WB, Lin WZ, Sheu ML. IL-13 downregulates PPAR-gamma/heme oxygenase-1 via ER stress-stimulated calpain activation: aggravation of activated microglia death. Cell Mol Life Sci. 2010;67:1465–1476. doi: 10.1007/s00018-009-0255-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Lo Verme J, Fu J, Astarita G, La Rana G, Russo R, Calignano A, Piomelli D. The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol. 2005;67:15–19. doi: 10.1124/mol.104.006353. [DOI] [PubMed] [Google Scholar]
  257. Losy J, Zaremba J, Skrobanski P. CXCL1 (GRO-alpha) chemokine in acute ischaemic stroke patients. Folia Neuropathol. 2005;43:97–102. [PubMed] [Google Scholar]
  258. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–341. doi: 10.1038/nature14432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Lu CJ, Sun Y, Muo CH, Chen RC, Chen PC, Hsu CY. Risk of stroke with thiazolidinediones: a ten-year nationwide population-based cohort study. Cerebrovasc Dis. 2013;36:145–151. doi: 10.1159/000353679. [DOI] [PubMed] [Google Scholar]
  260. Luna-Medina R, Cortes-Canteli M, Alonso M, Santos A, Martinez A, Perez-Castillo A. Regulation of inflammatory response in neural cells in vitro by thiadiazolidinones derivatives through peroxisome proliferator-activated receptor gamma activation. J Biol Chem. 2005;280:21453–21462. doi: 10.1074/jbc.M414390200. [DOI] [PubMed] [Google Scholar]
  261. Luo Y, Yin W, Signore AP, Zhang F, Hong Z, Wang S, Graham SH, Chen J. Neuroprotection against focal ischemic brain injury by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. J Neurochem. 2006;97:435–448. doi: 10.1111/j.1471-4159.2006.03758.x. [DOI] [PubMed] [Google Scholar]
  262. Luzzati F, De Marchis S, Fasolo A, Peretto P. Neurogenesis in the caudate nucleus of the adult rabbit. J Neurosci. 2006;26:609–621. doi: 10.1523/JNEUROSCI.4371-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Lygate CA, Hulbert K, Monfared M, Cole MA, Clarke K, Neubauer S. The PPARgamma-activator rosiglitazone does not alter remodeling but increases mortality in rats post-myocardial infarction. Cardiovasc Res. 2003;58:632–637. doi: 10.1016/s0008-6363(03)00289-x. [DOI] [PubMed] [Google Scholar]
  264. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J. 2013;166:240–249 e241. doi: 10.1016/j.ahj.2013.05.004. [DOI] [PubMed] [Google Scholar]
  265. Majai G, Sarang Z, Csomos K, Zahuczky G, Fesus L. PPARgamma-dependent regulation of human macrophages in phagocytosis of apoptotic cells. Eur J Immunol. 2007;37:1343–1354. doi: 10.1002/eji.200636398. [DOI] [PubMed] [Google Scholar]
  266. Mandai K, Matsumoto M, Kitagawa K, Matsushita K, Ohtsuki T, Mabuchi T, Colman DR, Kamada T, Yanagihara T. Ischemic damage and subsequent proliferation of oligodendrocytes in focal cerebral ischemia. Neuroscience. 1997;77:849–861. [PubMed] [Google Scholar]
  267. Mandrekar-Colucci S, Sauerbeck A, Popovich PG, McTigue DM. PPAR agonists as therapeutics for CNS trauma and neurological diseases. ASN Neuro. 2013;5:e00129. doi: 10.1042/AN20130030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Marklund N, Clausen F, Lewander T, Hillered L. Monitoring of reactive oxygen species production after traumatic brain injury in rats with microdialysis and the 4-hydroxybenzoic acid trapping method. J Neurotrauma. 2001;18:1217–1227. doi: 10.1089/089771501317095250. [DOI] [PubMed] [Google Scholar]
  269. Marx N, Mach F, Sauty A, Leung JH, Sarafi MN, Ransohoff RM, Libby P, Plutzky J, Luster AD. Peroxisome proliferator-activated receptor-gamma activators inhibit IFN-gamma-induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cells. J Immunol. 2000;164:6503–6508. doi: 10.4049/jimmunol.164.12.6503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Masuda T, Isobe Y, Aihara N, Furuyama F, Misumi S, Kim TS, Nishino H, Hida H. Increase in neurogenesis and neuroblast migration after a small intracerebral hemorrhage in rats. Neurosci Lett. 2007;425:114–119. doi: 10.1016/j.neulet.2007.08.039. [DOI] [PubMed] [Google Scholar]
  271. Matute C, Alberdi E, Domercq M, Sanchez-Gomez MV, Perez-Samartin A, Rodriguez-Antiguedad A, Perez-Cerda F. Excitotoxic damage to white matter. J Anat. 2007;210:693–702. doi: 10.1111/j.1469-7580.2007.00733.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. McGuire EJ, Woodside JR, Borden TA, Weiss RM. Prognostic value of urodynamic testing in myelodysplastic patients. J Urol. 1981;126:205–209. doi: 10.1016/s0022-5347(17)54449-3. [DOI] [PubMed] [Google Scholar]
  273. McKee CA, Lukens JR. Emerging Roles for the Immune System in Traumatic Brain Injury. Front Immunol. 2016;7:556. doi: 10.3389/fimmu.2016.00556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. McTigue DM. Potential Therapeutic Targets for PPARgamma after Spinal Cord Injury. PPAR Res. 2008;2008:517162. doi: 10.1155/2008/517162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. McTigue DM, Tripathi R, Wei P, Lash AT. The PPAR gamma agonist Pioglitazone improves anatomical and locomotor recovery after rodent spinal cord injury. Exp Neurol. 2007;205:396–406. doi: 10.1016/j.expneurol.2007.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Meletis K, Barnabe-Heider F, Carlen M, Evergren E, Tomilin N, Shupliakov O, Frisen J. Spinal cord injury reveals multilineage differentiation of ependymal cells. PLoS Biol. 2008;6:e182. doi: 10.1371/journal.pbio.0060182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Meng B, Zhang Q, Huang C, Zhang HT, Tang T, Yang HL. Effects of a single dose of methylprednisolone versus three doses of rosiglitazone on nerve growth factor levels after spinal cord injury. J Int Med Res. 2011;39:805–814. doi: 10.1177/147323001103900313. [DOI] [PubMed] [Google Scholar]
  278. Michalik L, Wahli W. Involvement of PPAR nuclear receptors in tissue injury and wound repair. J Clin Invest. 2006;116:598–606. doi: 10.1172/JCI27958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Mierzwa AJ, Marion CM, Sullivan GM, McDaniel DP, Armstrong RC. Components of myelin damage and repair in the progression of white matter pathology after mild traumatic brain injury. J Neuropathol Exp Neurol. 2015;74:218–232. doi: 10.1097/NEN.0000000000000165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Migaud M, Batailler M, Segura S, Duittoz A, Franceschini I, Pillon D. Emerging new sites for adult neurogenesis in the mammalian brain: a comparative study between the hypothalamus and the classical neurogenic zones. Eur J Neurosci. 2010;32:2042–2052. doi: 10.1111/j.1460-9568.2010.07521.x. [DOI] [PubMed] [Google Scholar]
  281. Min LJ, Mogi M, Shudou M, Jing F, Tsukuda K, Ohshima K, Iwanami J, Horiuchi M. Peroxisome proliferator- activated receptor-gamma activation with angiotensin II type 1 receptor blockade is pivotal for the prevention of blood-brain barrier impairment and cognitive decline in type 2 diabetic mice. Hypertension. 2012;59:1079–1088. doi: 10.1161/HYPERTENSIONAHA.112.192401. [DOI] [PubMed] [Google Scholar]
  282. Ming X, Ding M, Zhai B, Xiao L, Piao T, Liu M. Biochanin A inhibits lipopolysaccharide-induced inflammation in human umbilical vein endothelial cells. Life Sci. 2015;136:36–41. doi: 10.1016/j.lfs.2015.06.015. [DOI] [PubMed] [Google Scholar]
  283. Miron VE, Rajasekharan S, Jarjour AA, Zamvil SS, Kennedy TE, Antel JP. Simvastatin regulates oligodendroglial process dynamics and survival. Glia. 2007;55:130–143. doi: 10.1002/glia.20441. [DOI] [PubMed] [Google Scholar]
  284. Miron VE, Zehntner SP, Kuhlmann T, Ludwin SK, Owens T, Kennedy TE, Bedell BJ, Antel JP. Statin therapy inhibits remyelination in the central nervous system. Am J Pathol. 2009;174:1880–1890. doi: 10.2353/ajpath.2009.080947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Molina-Holgado E, Molina-Holgado F. Mending the broken brain: neuroimmune interactions in neurogenesis. J Neurochem. 2010;114:1277–1290. doi: 10.1111/j.1471-4159.2010.06849.x. [DOI] [PubMed] [Google Scholar]
  286. Molshatzki N, Orion D, Tsabari R, Schwammenthal Y, Merzeliak O, Toashi M, Tanne D. Chronic kidney disease in patients with acute intracerebral hemorrhage: association with large hematoma volume and poor outcome. Cerebrovasc Dis. 2011;31:271–277. doi: 10.1159/000322155. [DOI] [PubMed] [Google Scholar]
  287. Moreno S, Farioli-Vecchioli S, Ceru MP. Immunolocalization of peroxisome proliferator-activated receptors and retinoid X receptors in the adult rat CNS. Neuroscience. 2004;123:131–145. doi: 10.1016/j.neuroscience.2003.08.064. [DOI] [PubMed] [Google Scholar]
  288. Morganti-Kossmann MC, Rancan M, Otto VI, Stahel PF, Kossmann T. Role of cerebral inflammation after traumatic brain injury: a revisited concept. Shock. 2001;16:165–177. doi: 10.1097/00024382-200116030-00001. [DOI] [PubMed] [Google Scholar]
  289. Mracsko E, Liesz A, Stojanovic A, Lou WP, Osswald M, Zhou W, Karcher S, Winkler F, Martin-Villalba A, Cerwenka A, Veltkamp R. Antigen dependently activated cluster of differentiation 8-positive T cells cause perforin-mediated neurotoxicity in experimental stroke. J Neurosci. 2014;34:16784–16795. doi: 10.1523/JNEUROSCI.1867-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Mughal MM, Khan MK, DeMarco JK, Majid A, Shamoun F, Abela GS. Symptomatic and asymptomatic carotid artery plaque. Expert Rev Cardiovasc Ther. 2011;9:1315–1330. doi: 10.1586/erc.11.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Mukohda M, Stump M, Ketsawatsomkron P, Hu C, Quelle FW, Sigmund CD. Endothelial PPAR-gamma provides vascular protection from IL-1 beta-induced oxidative stress. Am J Physiol Heart Circ Physiol. 2016;310:H39–48. doi: 10.1152/ajpheart.00490.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Murray CJ, Lopez AD. Alternative projections of mortality and disability by cause 1990-2020: Global Burden of Disease Study. Lancet. 1997;349:1498–1504. doi: 10.1016/S0140-6736(96)07492-2. [DOI] [PubMed] [Google Scholar]
  293. Nakayama H, Jorgensen HS, Raaschou HO, Olsen TS. Recovery of upper extremity function in stroke patients: the Copenhagen Stroke Study. Arch Phys Med Rehabil. 1994;75:394–398. doi: 10.1016/0003-9993(94)90161-9. [DOI] [PubMed] [Google Scholar]
  294. Nathan DM, Buse JB, Davidson MB, Ferrannini E, Holman RR, Sherwin R, Zinman B, American Diabetes, A. European Association for Study of, D Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care. 2009;32:193–203. doi: 10.2337/dc08-9025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Neschen S, Morino K, Dong J, Wang-Fischer Y, Cline GW, Romanelli AJ, Rossbacher JC, Moore IK, Regittnig W, Munoz DS, Kim JH, Shulman GI. n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes. 2007;56:1034–1041. doi: 10.2337/db06-1206. [DOI] [PubMed] [Google Scholar]
  296. Neumann J, Sauerzweig S, Ronicke R, Gunzer F, Dinkel K, Ullrich O, Gunzer M, Reymann KG. Microglia cells protect neurons by direct engulfment of invading neutrophil granulocytes: a new mechanism of CNS immune privilege. J Neurosci. 2008;28:5965–5975. doi: 10.1523/JNEUROSCI.0060-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Ni W, Mao S, Xi G, Keep RF, Hua Y. Role of Erythrocyte CD47 in Intracerebral Hematoma Clearance. Stroke. 2016;47:505–511. doi: 10.1161/STROKEAHA.115.010920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Niino M, Iwabuchi K, Kikuchi S, Ato M, Morohashi T, Ogata A, Tashiro K, Onoe K. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by an agonist of peroxisome proliferator-activated receptor-gamma. J Neuroimmunol. 2001;116:40–48. doi: 10.1016/s0165-5728(01)00285-5. [DOI] [PubMed] [Google Scholar]
  299. Nissen SE, Wolski K. Rosiglitazone revisited: an updated meta-analysis of risk for myocardial infarction and cardiovascular mortality. Arch Intern Med. 2010;170:1191–1201. doi: 10.1001/archinternmed.2010.207. [DOI] [PubMed] [Google Scholar]
  300. Okada T, Wada J, Hida K, Eguchi J, Hashimoto I, Baba M, Yasuhara A, Shikata K, Makino H. Thiazolidinediones ameliorate diabetic nephropathy via cell cycle-dependent mechanisms. Diabetes. 2006;55:1666–1677. doi: 10.2337/db05-1285. [DOI] [PubMed] [Google Scholar]
  301. Ormerod BK, Hanft SJ, Asokan A, Haditsch U, Lee SW, Palmer TD. PPARgamma activation prevents impairments in spatial memory and neurogenesis following transient illness. Brain Behav Immun. 2013;29:28–38. doi: 10.1016/j.bbi.2012.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology. 1999;53:1937–1942. doi: 10.1212/wnl.53.9.1937. [DOI] [PubMed] [Google Scholar]
  303. Ou Z, Zhao X, Labiche LA, Strong R, Grotta JC, Herrmann O, Aronowski J. Neuronal expression of peroxisome proliferator-activated receptor-gamma (PPARgamma) and 15d-prostaglandin J2–mediated protection of brain after experimental cerebral ischemia in rat. Brain Res. 2006;1096:196–203. doi: 10.1016/j.brainres.2006.04.062. [DOI] [PubMed] [Google Scholar]
  304. Ouyang YB, Xu L, Liu S, Giffard RG. Role of Astrocytes in Delayed Neuronal Death: GLT-1 and its Novel Regulation by MicroRNAs. Adv Neurobiol. 2014;11:171–188. doi: 10.1007/978-3-319-08894-5_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Owens J, Wyper DJ, Patterson J, Brown DR, Elliott AT, Teasdale GM, McCulloch J. First SPET images of glutamate (NMDA) receptor activation in vivo in cerebral ischaemia. Nucl Med Commun. 1997;18:149–158. doi: 10.1097/00006231-199702000-00010. [DOI] [PubMed] [Google Scholar]
  306. Oyinbo CA. Secondary injury mechanisms in traumatic spinal cord injury: a nugget of this multiply cascade. Acta Neurobiol Exp (Wars) 2011;71:281–299. doi: 10.55782/ane-2011-1848. [DOI] [PubMed] [Google Scholar]
  307. Padilla J, Kaur K, Cao HJ, Smith TJ, Phipps RP. Peroxisome proliferator activator receptor-gamma agonists and 15-deoxy-Delta(12,14)(12,14)-PGJ(2) induce apoptosis in normal and malignant B-lineage cells. J Immunol. 2000;165:6941–6948. doi: 10.4049/jimmunol.165.12.6941. [DOI] [PubMed] [Google Scholar]
  308. Paintlia AS, Paintlia MK, Khan M, Vollmer T, Singh AK, Singh I. HMG-CoA reductase inhibitor augments survival and differentiation of oligodendrocyte progenitors in animal model of multiple sclerosis. FASEB J. 2005;19:1407–1421. doi: 10.1096/fj.05-3861com. [DOI] [PubMed] [Google Scholar]
  309. Paintlia AS, Paintlia MK, Singh AK, Orak JK, Singh I. Activation of PPAR-gamma and PTEN cascade participates in lovastatin-mediated accelerated differentiation of oligodendrocyte progenitor cells. Glia. 2010;58:1669–1685. doi: 10.1002/glia.21039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  310. Paintlia AS, Paintlia MK, Singh AK, Singh I. Inhibition of rho family functions by lovastatin promotes myelin repair in ameliorating experimental autoimmune encephalomyelitis. Mol Pharmacol. 2008;73:1381–1393. doi: 10.1124/mol.107.044230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Pan HC, Yang CN, Hung YW, Lee WJ, Tien HR, Shen CC, Sheehan J, Chou CT, Sheu ML. Reciprocal modulation of C/EBP-alpha and C/EBP-beta by IL-13 in activated microglia prevents neuronal death. Eur J Immunol. 2013;43:2854–2865. doi: 10.1002/eji.201343301. [DOI] [PubMed] [Google Scholar]
  312. Pang Y, Cai Z, Rhodes PG. Effect of tumor necrosis factor-alpha on developing optic nerve oligodendrocytes in culture. J Neurosci Res. 2005;80:226–234. doi: 10.1002/jnr.20450. [DOI] [PubMed] [Google Scholar]
  313. Pantoni L, Garcia JH, Gutierrez JA. Cerebral white matter is highly vulnerable to ischemia. Stroke. 1996;27:1641–1646. doi: 10.1161/01.str.27.9.1641. discussion 1647. [DOI] [PubMed] [Google Scholar]
  314. Park BV, Pan F. The role of nuclear receptors in regulation of Th17/Treg biology and its implications for diseases. Cell Mol Immunol. 2015;12:533–542. doi: 10.1038/cmi.2015.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Park EJ, Park SY, Joe EH, Jou I. 15d-PGJ2 and rosiglitazone suppress Janus kinase-STAT inflammatory signaling through induction of suppressor of cytokine signaling 1 (SOCS1) and SOCS3 in glia. J Biol Chem. 2003;278:14747–14752. doi: 10.1074/jbc.M210819200. [DOI] [PubMed] [Google Scholar]
  316. Park EY, Cho IJ, Kim SG. Transactivation of the PPAR-responsive enhancer module in chemopreventive glutathione S-transferase gene by the peroxisome proliferator-activated receptor-gamma and retinoid X receptor heterodimer. Cancer Res. 2004a;64:3701–3713. doi: 10.1158/0008-5472.CAN-03-3924. [DOI] [PubMed] [Google Scholar]
  317. Park KS, Lee RD, Kang SK, Han SY, Park KL, Yang KH, Song YS, Park HJ, Lee YM, Yun YP, Oh KW, Kim DJ, Yun YW, Hwang SJ, Lee SE, Hong JT. Neuronal differentiation of embryonic midbrain cells by upregulation of peroxisome proliferator-activated receptor-gamma via the JNK-dependent pathway. Exp Cell Res. 2004b;297:424–433. doi: 10.1016/j.yexcr.2004.03.034. [DOI] [PubMed] [Google Scholar]
  318. Pasceri V, Wu HD, Willerson JT, Yeh ET. Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor-gamma activators. Circulation. 2000;101:235–238. doi: 10.1161/01.cir.101.3.235. [DOI] [PubMed] [Google Scholar]
  319. Pascual G, Fong AL, Ogawa S, Gamliel A, Li AC, Perissi V, Rose DW, Willson TM, Rosenfeld MG, Glass CK. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature. 2005;437:759–763. doi: 10.1038/nature03988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Patel AR, Ritzel R, McCullough LD, Liu F. Microglia and ischemic stroke: a double-edged sword. Int J Physiol Pathophysiol Pharmacol. 2013;5:73–90. [PMC free article] [PubMed] [Google Scholar]
  321. Patel PC, Harrison RE. Membrane ruffles capture C3bi-opsonized particles in activated macrophages. Mol Biol Cell. 2008;19:4628–4639. doi: 10.1091/mbc.E08-02-0223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Patel SN, Serghides L, Smith TG, Febbraio M, Silverstein RL, Kurtz TW, Pravenec M, Kain KC. CD36 mediates the phagocytosis of Plasmodium falciparum-infected erythrocytes by rodent macrophages. J Infect Dis. 2004;189:204–213. doi: 10.1086/380764. [DOI] [PubMed] [Google Scholar]
  323. Paterniti I, Impellizzeri D, Crupi R, Morabito R, Campolo M, Esposito E, Cuzzocrea S. Molecular evidence for the involvement of PPAR-delta and PPAR-gamma in anti-inflammatory and neuroprotective activities of palmitoylethanolamide after spinal cord trauma. J Neuroinflammation. 2013;10:20. doi: 10.1186/1742-2094-10-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol. 2006;199:265–273. doi: 10.1016/j.expneurol.2006.01.018. [DOI] [PubMed] [Google Scholar]
  325. Pennypacker KR, Offner H. The role of the spleen in ischemic stroke. J Cereb Blood Flow Metab. 2015;35:186–187. doi: 10.1038/jcbfm.2014.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Piantadosi CA, Zhang J. Mitochondrial generation of reactive oxygen species after brain ischemia in the rat. Stroke. 1996;27:327–331. doi: 10.1161/01.str.27.2.327. discussion 332. [DOI] [PubMed] [Google Scholar]
  327. Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, Machado De Oliveira R, Leid M, McBurney MW, Guarente L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature. 2004;429:771–776. doi: 10.1038/nature02583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  328. Pierce AA, Xu AW. De novo neurogenesis in adult hypothalamus as a compensatory mechanism to regulate energy balance. J Neurosci. 2010;30:723–730. doi: 10.1523/JNEUROSCI.2479-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Pittet JF, Lee H, Morabito D, Howard MB, Welch WJ, Mackersie RC. Serum levels of Hsp 72 measured early after trauma correlate with survival. J Trauma. 2002;52:611–617. doi: 10.1097/00005373-200204000-00001. discussion 617. [DOI] [PubMed] [Google Scholar]
  330. Polvani S, Tarocchi M, Galli A. PPARgamma and Oxidative Stress: Con(beta) Catenating NRF2 and FOXO. PPAR Res. 2012;2012:641087. doi: 10.1155/2012/641087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  331. Pourcet B, Pineda-Torra I, Derudas B, Staels B, Glineur C. SUMOylation of human peroxisome proliferator-activated receptor alpha inhibits its trans-activity through the recruitment of the nuclear corepressor NCoR. J Biol Chem. 2010;285:5983–5992. doi: 10.1074/jbc.M109.078311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Puyal J, Ginet V, Clarke PG. Multiple interacting cell death mechanisms in the mediation of excitotoxicity and ischemic brain damage: a challenge for neuroprotection. Prog Neurobiol. 2013;105:24–48. doi: 10.1016/j.pneurobio.2013.03.002. [DOI] [PubMed] [Google Scholar]
  333. Qi L, Jacob A, Wang P, Wu R. Peroxisome proliferator activated receptor-gamma and traumatic brain injury. Int J Clin Exp Med. 2010;3:283–292. [PMC free article] [PubMed] [Google Scholar]
  334. Quinones-Hinojosa A, Sanai N, Soriano-Navarro M, Gonzalez-Perez O, Mirzadeh Z, Gil-Perotin S, Romero-Rodriguez R, Berger MS, Garcia-Verdugo JM, Alvarez-Buylla A. Cellular composition and cytoarchitecture of the adult human subventricular zone: a niche of neural stem cells. J Comp Neurol. 2006;494:415–434. doi: 10.1002/cne.20798. [DOI] [PubMed] [Google Scholar]
  335. Qureshi AI, Mendelow AD, Hanley DF. Intracerebral haemorrhage. Lancet. 2009;373:1632–1644. doi: 10.1016/S0140-6736(09)60371-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  336. Qureshi AI, Qureshi MH. Acute hypertensive response in patients with intracerebral hemorrhage pathophysiology and treatment. J Cereb Blood Flow Metab. 2017:271678X17725431. doi: 10.1177/0271678X17725431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Qureshi AI, Tuhrim S, Broderick JP, Batjer HH, Hondo H, Hanley DF. Spontaneous intracerebral hemorrhage. N Engl J Med. 2001;344:1450–1460. doi: 10.1056/NEJM200105103441907. [DOI] [PubMed] [Google Scholar]
  338. Ramirez-Amaya V, Marrone DF, Gage FH, Worley PF, Barnes CA. Integration of new neurons into functional neural networks. J Neurosci. 2006;26:12237–12241. doi: 10.1523/JNEUROSCI.2195-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Randall RD, Thayer SA. Glutamate-induced calcium transient triggers delayed calcium overload and neurotoxicity in rat hippocampal neurons. J Neurosci. 1992;12:1882–1895. doi: 10.1523/JNEUROSCI.12-05-01882.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Redondo-Castro E, Hernandez J, Mahy N, Navarro X. Phagocytic microglial phenotype induced by glibenclamide improves functional recovery but worsens hyperalgesia after spinal cord injury in adult rats. Eur J Neurosci. 2013;38:3786–3798. doi: 10.1111/ejn.12382. [DOI] [PubMed] [Google Scholar]
  341. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998;391:79–82. doi: 10.1038/34178. [DOI] [PubMed] [Google Scholar]
  342. Riyahi TY, Frese F, Steinert M, Omosigho NN, Glockner G, Eichinger L, Orabi B, Williams RS, Noegel AA. RpkA, a highly conserved GPCR with a lipid kinase domain, has a role in phagocytosis and anti-bacterial defense. PLoS One. 2011;6:e27311. doi: 10.1371/journal.pone.0027311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Robinson MB. The family of sodium-dependent glutamate transporters: a focus on the GLT-1/EAAT2 subtype. Neurochem Int. 1998;33:479–491. doi: 10.1016/s0197-0186(98)00055-2. [DOI] [PubMed] [Google Scholar]
  344. Rohatagi S, Carrothers TJ, Jin J, Jusko WJ, Khariton T, Walker J, Truitt K, Salazar DE. Model-based development of a PPARgamma agonist, rivoglitazone, to aid dose selection and optimize clinical trial designs. J Clin Pharmacol. 2008;48:1420–1429. doi: 10.1177/0091270008323260. [DOI] [PubMed] [Google Scholar]
  345. Rolland M, Li X, Sellier Y, Martin H, Perez-Berezo T, Rauwel B, Benchoua A, Bessieres B, Aziza J, Cenac N, Luo M, Casper C, Peschanski M, Gonzalez-Dunia D, Leruez-Ville M, Davrinche C, Chavanas S. PPARgamma Is Activated during Congenital Cytomegalovirus Infection and Inhibits Neuronogenesis from Human Neural Stem Cells. PLoS Pathog. 2016;12:e1005547. doi: 10.1371/journal.ppat.1005547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Romera C, Hurtado O, Mallolas J, Pereira MP, Morales JR, Romera A, Serena J, Vivancos J, Nombela F, Lorenzo P, Lizasoain I, Moro MA. Ischemic preconditioning reveals that GLT1/EAAT2 glutamate transporter is a novel PPARgamma target gene involved in neuroprotection. J Cereb Blood Flow Metab. 2007;27:1327–1338. doi: 10.1038/sj.jcbfm.9600438. [DOI] [PubMed] [Google Scholar]
  347. Rosenberg GA. Inflammation and white matter damage in vascular cognitive impairment. Stroke. 2009;40:S20–23. doi: 10.1161/STROKEAHA.108.533133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Rosenberg GA. Vascular cognitive impairment: Biomarkers in diagnosis and molecular targets in therapy. J Cereb Blood Flow Metab. 2016;36:4–5. doi: 10.1177/0271678X15609542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Rosenbluth J. Redundant myelin sheaths and other ultrastructural features of the toad cerebellum. J Cell Biol. 1966;28:73–93. doi: 10.1083/jcb.28.1.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Ross GD, Reed W, Dalzell JG, Becker SE, Hogg N. Macrophage cytoskeleton association with CR3 and CR4 regulates receptor mobility and phagocytosis of iC3b-opsonized erythrocytes. J Leukoc Biol. 1992;51:109–117. doi: 10.1002/jlb.51.2.109. [DOI] [PubMed] [Google Scholar]
  351. Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature. 1993;362:801–809. doi: 10.1038/362801a0. [DOI] [PubMed] [Google Scholar]
  352. Ross R. Atherosclerosis–an inflammatory disease. N Engl J Med. 1999;340:115–126. doi: 10.1056/NEJM199901143400207. [DOI] [PubMed] [Google Scholar]
  353. Roth AD, Leisewitz AV, Jung JE, Cassina P, Barbeito L, Inestrosa NC, Bronfman M. PPAR gamma activators induce growth arrest and process extension in B12 oligodendrocyte-like cells and terminal differentiation of cultured oligodendrocytes. J Neurosci Res. 2003;72:425–435. doi: 10.1002/jnr.10596. [DOI] [PubMed] [Google Scholar]
  354. Rotshenker S. Microglia and macrophage activation and the regulation of complement-receptor-3 (CR3/MAC-1)-mediated myelin phagocytosis in injury and disease. J Mol Neurosci. 2003;21:65–72. doi: 10.1385/JMN:21:1:65. [DOI] [PubMed] [Google Scholar]
  355. Rotshenker S. The role of Galectin-3/MAC-2 in the activation of the innate-immune function of phagocytosis in microglia in injury and disease. J Mol Neurosci. 2009;39:99–103. doi: 10.1007/s12031-009-9186-7. [DOI] [PubMed] [Google Scholar]
  356. Rowat A, Graham C, Dennis M. Renal dysfunction in stroke patients: a hospital-based cohort study and systematic review. Int J Stroke. 2014;9:633–639. doi: 10.1111/ijs.12264. [DOI] [PubMed] [Google Scholar]
  357. Rushmore TH, Morton MR, Pickett CB. The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity. J Biol Chem. 1991;266:11632–11639. [PubMed] [Google Scholar]
  358. Rushmore TH, Pickett CB. Transcriptional regulation of the rat glutathione S-transferase Ya subunit gene. Characterization of a xenobiotic-responsive element controlling inducible expression by phenolic antioxidants. J Biol Chem. 1990;265:14648–14653. [PubMed] [Google Scholar]
  359. Sahay A, Scobie KN, Hill AS, O’Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R. Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature. 2011;472:466–470. doi: 10.1038/nature09817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Salehi A, Zhang JH, Obenaus A. Response of the cerebral vasculature following traumatic brain injury. J Cereb Blood Flow Metab. 2017;37:2320–2339. doi: 10.1177/0271678X17701460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Salter MG, Fern R. NMDA receptors are expressed in developing oligodendrocyte processes and mediate injury. Nature. 2005;438:1167–1171. doi: 10.1038/nature04301. [DOI] [PubMed] [Google Scholar]
  362. Saluja I, Granneman JG, Skoff RP. PPAR delta agonists stimulate oligodendrocyte differentiation in tissue culture. Glia. 2001;33:191–204. [PubMed] [Google Scholar]
  363. Samaha FF, Szapary PO, Iqbal N, Williams MM, Bloedon LT, Kochar A, Wolfe ML, Rader DJ. Effects of rosiglitazone on lipids, adipokines, and inflammatory markers in nondiabetic patients with low high-density lipoprotein cholesterol and metabolic syndrome. Arterioscler Thromb Vasc Biol. 2006;26:624–630. doi: 10.1161/01.ATV.0000200136.56716.30. [DOI] [PubMed] [Google Scholar]
  364. Sarafidis PA, Lasaridis AN. Actions of peroxisome proliferator-activated receptors-gamma agonists explaining a possible blood pressure-lowering effect. Am J Hypertens. 2006;19:646–653. doi: 10.1016/j.amjhyper.2005.12.017. [DOI] [PubMed] [Google Scholar]
  365. Sarafidis PA, Stafylas PC, Georgianos PI, Saratzis AN, Lasaridis AN. Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis. Am J Kidney Dis. 2010;55:835–847. doi: 10.1053/j.ajkd.2009.11.013. [DOI] [PubMed] [Google Scholar]
  366. Sasaki M, Jordan P, Welbourne T, Minagar A, Joh T, Itoh M, Elrod JW, Alexander JS. Troglitazone, a PPAR-gamma activator prevents endothelial cell adhesion molecule expression and lymphocyte adhesion mediated by TNF-alpha. BMC Physiol. 2005;5:3. doi: 10.1186/1472-6793-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  367. Sauer S. Ligands for the Nuclear Peroxisome Proliferator-Activated Receptor Gamma. Trends Pharmacol Sci. 2015;36:688–704. doi: 10.1016/j.tips.2015.06.010. [DOI] [PubMed] [Google Scholar]
  368. Scheen AJ. Outcomes and lessons from the PROactive study. Diabetes Res Clin Pract. 2012;98:175–186. doi: 10.1016/j.diabres.2012.09.001. [DOI] [PubMed] [Google Scholar]
  369. Schilling M, Besselmann M, Muller M, Strecker JK, Ringelstein EB, Kiefer R. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: an investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2005;196:290–297. doi: 10.1016/j.expneurol.2005.08.004. [DOI] [PubMed] [Google Scholar]
  370. Schirmer-Mikalsen K, Vik A, Gisvold SE, Skandsen T, Hynne H, Klepstad P. Severe head injury: control of physiological variables, organ failure and complications in the intensive care unit. Acta Anaesthesiol Scand. 2007;51:1194–1201. doi: 10.1111/j.1399-6576.2007.01372.x. [DOI] [PubMed] [Google Scholar]
  371. Schousboe A, Waagepetersen HS. Role of astrocytes in glutamate homeostasis: implications for excitotoxicity. Neurotox Res. 2005;8:221–225. doi: 10.1007/BF03033975. [DOI] [PubMed] [Google Scholar]
  372. Schulz MK, Wang LP, Tange M, Bjerre P. Cerebral microdialysis monitoring: determination of normal and ischemic cerebral metabolisms in patients with aneurysmal subarachnoid hemorrhage. J Neurosurg. 2000;93:808–814. doi: 10.3171/jns.2000.93.5.0808. [DOI] [PubMed] [Google Scholar]
  373. Semple RK, Chatterjee VK, O’Rahilly S. PPAR gamma and human metabolic disease. J Clin Invest. 2006;116:581–589. doi: 10.1172/JCI28003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  374. Seri B, Garcia-Verdugo JM, McEwen BS, Alvarez-Buylla A. Astrocytes give rise to new neurons in the adult mammalian hippocampus. J Neurosci. 2001;21:7153–7160. doi: 10.1523/JNEUROSCI.21-18-07153.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Shao D, Rangwala SM, Bailey ST, Krakow SL, Reginato MJ, Lazar MA. Interdomain communication regulating ligand binding by PPAR-gamma. Nature. 1998;396:377–380. doi: 10.1038/24634. [DOI] [PubMed] [Google Scholar]
  376. Sharma SK, Naidu G. The role of danger-associated molecular patterns (DAMPs) in trauma and infections. J Thorac Dis. 2016;8:1406–1409. doi: 10.21037/jtd.2016.05.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Shattil SJ, Kim C, Ginsberg MH. The final steps of integrin activation: the end game. Nat Rev Mol Cell Biol. 2010;11:288–300. doi: 10.1038/nrm2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Shaya FT, Lu Z, Sohn K, Weir MR. Thiazolidinediones and cardiovascular events in high-risk patients with type-2 diabetes mellitus: a comparison with other oral antidiabetic agents. P T. 2009;34:490–501. [PMC free article] [PubMed] [Google Scholar]
  379. Shen J, Xie L, Mao X, Zhou Y, Zhan R, Greenberg DA, Jin K. Neurogenesis after primary intracerebral hemorrhage in adult human brain. J Cereb Blood Flow Metab. 2008;28:1460–1468. doi: 10.1038/jcbfm.2008.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  380. Shereen A, Nemkul N, Yang D, Adhami F, Dunn RS, Hazen ML, Nakafuku M, Ning G, Lindquist DM, Kuan CY. Ex vivo diffusion tensor imaging and neuropathological correlation in a murine model of hypoxia-ischemia-induced thrombotic stroke. J Cereb Blood Flow Metab. 2011;31:1155–1169. doi: 10.1038/jcbfm.2010.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636–645. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
  382. Shevach EM, DiPaolo RA, Andersson J, Zhao DM, Stephens GL, Thornton AM. The lifestyle of naturally occurring CD4+ CD25+ Foxp3+ regulatory T cells. Immunol Rev. 2006;212:60–73. doi: 10.1111/j.0105-2896.2006.00415.x. [DOI] [PubMed] [Google Scholar]
  383. Shi H, Hu X, Leak RK, Shi Y, An C, Suenaga J, Chen J, Gao Y. Demyelination as a rational therapeutic target for ischemic or traumatic brain injury. Exp Neurol. 2015;272:17–25. doi: 10.1016/j.expneurol.2015.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Shi Y, Leak RK, Keep RF, Chen J. Translational Stroke Research on Blood-Brain Barrier Damage: Challenges, Perspectives, and Goals. Transl Stroke Res. 2016a;7:89–92. doi: 10.1007/s12975-016-0447-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Shi Y, Zhang L, Pu H, Mao L, Hu X, Jiang X, Xu N, Stetler RA, Zhang F, Liu X, Leak RK, Keep RF, Ji X, Chen J. Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun. 2016b;7:10523. doi: 10.1038/ncomms10523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Shih AY, Imbeault S, Barakauskas V, Erb H, Jiang L, Li P, Murphy TH. Induction of the Nrf2-driven antioxidant response confers neuroprotection during mitochondrial stress in vivo. J Biol Chem. 2005;280:22925–22936. doi: 10.1074/jbc.M414635200. [DOI] [PubMed] [Google Scholar]
  387. Shiomi T, Tsutsui H, Hayashidani S, Suematsu N, Ikeuchi M, Wen J, Ishibashi M, Kubota T, Egashira K, Takeshita A. Pioglitazone, a peroxisome proliferator-activated receptor-gamma agonist, attenuates left ventricular remodeling and failure after experimental myocardial infarction. Circulation. 2002;106:3126–3132. doi: 10.1161/01.cir.0000039346.31538.2c. [DOI] [PubMed] [Google Scholar]
  388. Siddiqui AM, Cui X, Wu R, Dong W, Zhou M, Hu M, Simms HH, Wang P. The anti-inflammatory effect of curcumin in an experimental model of sepsis is mediated by up-regulation of peroxisome proliferator-activated receptor-gamma. Crit Care Med. 2006;34:1874–1882. doi: 10.1097/01.CCM.0000221921.71300.BF. [DOI] [PubMed] [Google Scholar]
  389. Sidell RJ, Cole MA, Draper NJ, Desrois M, Buckingham RE, Clarke K. Thiazolidinedione treatment normalizes insulin resistance and ischemic injury in the zucker Fatty rat heart. Diabetes. 2002;51:1110–1117. doi: 10.2337/diabetes.51.4.1110. [DOI] [PubMed] [Google Scholar]
  390. Sim FJ, Lang JK, Ali TA, Roy NS, Vates GE, Pilcher WH, Goldman SA. Statin treatment of adult human glial progenitors induces PPAR gamma-mediated oligodendrocytic differentiation. Glia. 2008;56:954–962. doi: 10.1002/glia.20669. [DOI] [PubMed] [Google Scholar]
  391. Sim FJ, Zhao C, Penderis J, Franklin RJ. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J Neurosci. 2002;22:2451–2459. doi: 10.1523/JNEUROSCI.22-07-02451.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Simoni J, Feola M, Canizaro PC. Generation of free oxygen radicals and the toxicity of hemoglobin solutions. Biomater Artif Cells Artif Organs. 1990;18:189–202. doi: 10.3109/10731199009117301. [DOI] [PubMed] [Google Scholar]
  393. Smith KJ, Blakemore WF, McDonald WI. Central remyelination restores secure conduction. Nature. 1979;280:395–396. doi: 10.1038/280395a0. [DOI] [PubMed] [Google Scholar]
  394. Soares HD, Hicks RR, Smith D, McIntosh TK. Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury. J Neurosci. 1995;15:8223–8233. doi: 10.1523/JNEUROSCI.15-12-08223.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Staessen JA, Kuznetsova T, Stolarz K. Hypertension prevalence and stroke mortality across populations. JAMA. 2003;289:2420–2422. doi: 10.1001/jama.289.18.2420. [DOI] [PubMed] [Google Scholar]
  396. Stephen RL, Gustafsson MC, Jarvis M, Tatoud R, Marshall BR, Knight D, Ehrenborg E, Harris AL, Wolf CR, Palmer CN. Activation of peroxisome proliferator-activated receptor delta stimulates the proliferation of human breast and prostate cancer cell lines. Cancer Res. 2004;64:3162–3170. doi: 10.1158/0008-5472.can-03-2760. [DOI] [PubMed] [Google Scholar]
  397. Storer PD, Xu J, Chavis JA, Drew PD. Cyclopentenone prostaglandins PGA2 and 15-deoxy-delta12, 14 PGJ2 suppress activation of murine microglia and astrocytes: implications for multiple sclerosis. J Neurosci Res. 2005;80:66–74. doi: 10.1002/jnr.20413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Streilein JW. Unraveling immune privilege. Science. 1995;270:1158–1159. doi: 10.1126/science.270.5239.1158. [DOI] [PubMed] [Google Scholar]
  399. Sugawara A, Takeuchi K, Uruno A, Ikeda Y, Arima S, Kudo M, Sato K, Taniyama Y, Ito S. Transcriptional suppression of type 1 angiotensin II receptor gene expression by peroxisome proliferator-activated receptor-gamma in vascular smooth muscle cells. Endocrinology. 2001;142:3125–3134. doi: 10.1210/endo.142.7.8272. [DOI] [PubMed] [Google Scholar]
  400. Sullivan GM, Mierzwa AJ, Kijpaisalratana N, Tang H, Wang Y, Song SK, Selwyn R, Armstrong RC. Oligodendrocyte lineage and subventricular zone response to traumatic axonal injury in the corpus callosum. J Neuropathol Exp Neurol. 2013;72:1106–1125. doi: 10.1097/NEN.0000000000000009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Sundararajan S, Landreth GE. Antiinflammatory properties of PPARgamma agonists following ischemia. Drug News Perspect. 2004;17:229–236. doi: 10.1358/dnp.2004.17.4.829049. [DOI] [PubMed] [Google Scholar]
  402. Taheri M, Salamian A, Ghaedi K, Peymani M, Izadi T, Nejati AS, Atefi A, Nematollahi M, Ahmadi Ghahrizjani F, Esmaeili M, Kiani Esfahani A, Irani S, Baharvand H, Nasr-Esfahani MH. A ground state of PPARgamma activity and expression is required for appropriate neural differentiation of hESCs. Pharmacol Rep. 2015;67:1103–1114. doi: 10.1016/j.pharep.2015.04.011. [DOI] [PubMed] [Google Scholar]
  403. Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, Sonobe Y, Mizuno T, Suzumura A. Tumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. J Biol Chem. 2006;281:21362–21368. doi: 10.1074/jbc.M600504200. [DOI] [PubMed] [Google Scholar]
  404. Tanaka R, Yamashiro K, Okuma Y, Shimura H, Nakamura S, Ueno Y, Tanaka Y, Miyamoto N, Tomizawa Y, Nakahara T, Furukawa Y, Watada H, Kawamori R, Hattori N, Urabe T. Effects of Pioglitazone for Secondary Stroke Prevention in Patients with Impaired Glucose Tolerance and Newly Diagnosed Diabetes: The J-SPIRIT Study. J Atheroscler Thromb. 2015;22:1305–1316. doi: 10.5551/jat.30007. [DOI] [PubMed] [Google Scholar]
  405. Tang Y, Le W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol Neurobiol. 2016;53:1181–1194. doi: 10.1007/s12035-014-9070-5. [DOI] [PubMed] [Google Scholar]
  406. Tekkok SB, Goldberg MP. Ampa/kainate receptor activation mediates hypoxic oligodendrocyte death and axonal injury in cerebral white matter. J Neurosci. 2001;21:4237–4248. doi: 10.1523/JNEUROSCI.21-12-04237.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Terasaki Y, Liu Y, Hayakawa K, Pham LD, Lo EH, Ji X, Arai K. Mechanisms of neurovascular dysfunction in acute ischemic brain. Curr Med Chem. 2014;21:2035–2042. doi: 10.2174/0929867321666131228223400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Tesfamariam B, DeFelice AF. Endothelial injury in the initiation and progression of vascular disorders. Vascul Pharmacol. 2007;46:229–237. doi: 10.1016/j.vph.2006.11.005. [DOI] [PubMed] [Google Scholar]
  409. Thal SC, Heinemann M, Luh C, Pieter D, Werner C, Engelhard K. Pioglitazone reduces secondary brain damage after experimental brain trauma by PPAR-gamma-independent mechanisms. J Neurotrauma. 2011;28:983–993. doi: 10.1089/neu.2010.1685. [DOI] [PubMed] [Google Scholar]
  410. Thal SC, Neuhaus W. The blood-brain barrier as a target in traumatic brain injury treatment. Arch Med Res. 2014;45:698–710. doi: 10.1016/j.arcmed.2014.11.006. [DOI] [PubMed] [Google Scholar]
  411. Thal SC, Wyschkon S, Pieter D, Engelhard K, Werner C. Selection of endogenous control genes for normalization of gene expression analysis after experimental brain trauma in mice. J Neurotrauma. 2008;25:785–794. doi: 10.1089/neu.2007.0497. [DOI] [PubMed] [Google Scholar]
  412. Tholance Y, Barcelos G, Dailler F, Perret-Liaudet A, Renaud B. Clinical Neurochemistry of Subarachnoid Hemorrhage: Toward Predicting Individual Outcomes via Biomarkers of Brain Energy Metabolism. ACS Chem Neurosci. 2015;6:1902–1905. doi: 10.1021/acschemneuro.5b00299. [DOI] [PubMed] [Google Scholar]
  413. Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 1998;93:241–252. doi: 10.1016/s0092-8674(00)81575-5. [DOI] [PubMed] [Google Scholar]
  414. Trang T, Beggs S, Salter MW. Brain-derived neurotrophic factor from microglia: a molecular substrate for neuropathic pain. Neuron Glia Biol. 2011;7:99–108. doi: 10.1017/S1740925X12000087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  415. Tripathi R, McTigue DM. Prominent oligodendrocyte genesis along the border of spinal contusion lesions. Glia. 2007;55:698–711. doi: 10.1002/glia.20491. [DOI] [PubMed] [Google Scholar]
  416. Tsai YS, Xu L, Smithies O, Maeda N. Genetic variations in peroxisome proliferator-activated receptor gamma expression affect blood pressure. Proc Natl Acad Sci U S A. 2009;106:19084–19089. doi: 10.1073/pnas.0909657106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Tymianski M, Charlton MP, Carlen PL, Tator CH. Secondary Ca2+ overload indicates early neuronal injury which precedes staining with viability indicators. Brain Res. 1993;607:319–323. doi: 10.1016/0006-8993(93)91523-u. [DOI] [PubMed] [Google Scholar]
  418. Urra X, Obach V, Chamorro A. Stroke induced immunodepression syndrome: from bench to bedside. Curr Mol Med. 2009;9:195–202. doi: 10.2174/156652409787581574. [DOI] [PubMed] [Google Scholar]
  419. Uruno A, Matsuda K, Noguchi N, Yoshikawa T, Kudo M, Satoh F, Rainey WE, Hui XG, Akahira J, Nakamura Y, Sasano H, Okamoto H, Ito S, Sugawara A. Peroxisome proliferator-activated receptor-{gamma} suppresses CYP11B2 expression and aldosterone production. J Mol Endocrinol. 2011;46:37–49. doi: 10.1677/JME-10-0088. [DOI] [PubMed] [Google Scholar]
  420. Vallarino C, Perez A, Fusco G, Liang H, Bron M, Manne S, Joseph G, Yu S. Comparing pioglitazone to insulin with respect to cancer, cardiovascular and bone fracture endpoints, using propensity score weights. Clin Drug Investig. 2013;33:621–631. doi: 10.1007/s40261-013-0106-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Vanasse A, Carpentier AC, Courteau J, Asghari S. Stroke and cardiovascular morbidity and mortality associated with rosiglitazone use in elderly diabetic patients. Diab Vasc Dis Res. 2009;6:87–93. doi: 10.1177/1479164109336047. [DOI] [PubMed] [Google Scholar]
  422. Vattulainen-Collanus S, Akinrinade O, Li M, Koskenvuo M, Li CG, Rao SP, de Jesus Perez V, Yuan K, Sawada H, Koskenvuo JW, Alvira C, Rabinovitch M, Alastalo TP. Loss of PPARgamma in endothelial cells leads to impaired angiogenesis. J Cell Sci. 2016;129:693–705. doi: 10.1242/jcs.169011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. Venkatesan C, Chrzaszcz M, Choi N, Wainwright MS. Chronic upregulation of activated microglia immunoreactive for galectin-3/Mac-2 and nerve growth factor following diffuse axonal injury. J Neuroinflammation. 2010;7:32. doi: 10.1186/1742-2094-7-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Verrier E, Wang L, Wadham C, Albanese N, Hahn C, Gamble JR, Chatterjee VK, Vadas MA, Xia P. PPARgamma agonists ameliorate endothelial cell activation via inhibition of diacylglycerol-protein kinase C signaling pathway: role of diacylglycerol kinase. Circ Res. 2004;94:1515–1522. doi: 10.1161/01.RES.0000130527.92537.06. [DOI] [PubMed] [Google Scholar]
  425. Victor NA, Wanderi EW, Gamboa J, Zhao X, Aronowski J, Deininger K, Lust WD, Landreth GE, Sundararajan S. Altered PPARgamma expression and activation after transient focal ischemia in rats. Eur J Neurosci. 2006;24:1653–1663. doi: 10.1111/j.1460-9568.2006.05037.x. [DOI] [PubMed] [Google Scholar]
  426. Viola HM, Hool LC. Role of the cytoskeleton in communication between L-type Ca(2+) channels and mitochondria. Clin Exp Pharmacol Physiol. 2013;40:295–304. doi: 10.1111/1440-1681.12072. [DOI] [PubMed] [Google Scholar]
  427. Viscoli CM, Brass LM, Carolei A, Conwit R, Ford GA, Furie KL, Gorman M, Guarino PD, Inzucchi SE, Lovejoy AM, Parsons MW, Peduzzi PN, Ringleb PA, Schwartz GG, Spence JD, Tanne D, Young LH, Kernan WN, investigators, I.T. Pioglitazone for secondary prevention after ischemic stroke and transient ischemic attack: rationale and design of the Insulin Resistance Intervention after Stroke Trial. Am Heart J. 2014;168:823–829 e826. doi: 10.1016/j.ahj.2014.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Von Knethen AA, Brune B. Delayed activation of PPARgamma by LPS and IFN-gamma attenuates the oxidative burst in macrophages. FASEB J. 2001;15:535–544. doi: 10.1096/fj.00-0187com. [DOI] [PubMed] [Google Scholar]
  429. Wada K, Nakajima A, Katayama K, Kudo C, Shibuya A, Kubota N, Terauchi Y, Tachibana M, Miyoshi H, Kamisaki Y, Mayumi T, Kadowaki T, Blumberg RS. Peroxisome proliferator-activated receptor gamma-mediated regulation of neural stem cell proliferation and differentiation. J Biol Chem. 2006;281:12673–12681. doi: 10.1074/jbc.M513786200. [DOI] [PubMed] [Google Scholar]
  430. Wahl LM, Shankavaram U, Zhang Y. Role of macrophages in vascular tissue remodelling. Transpl Immunol. 1997;5:173–176. doi: 10.1016/s0966-3274(97)80033-0. [DOI] [PubMed] [Google Scholar]
  431. Waite KJ, Floyd ZE, Arbour-Reily P, Stephens JM. Interferon-gamma-induced regulation of peroxisome proliferator- activated receptor gamma and STATs in adipocytes. J Biol Chem. 2001;276:7062–7068. doi: 10.1074/jbc.M007894200. [DOI] [PubMed] [Google Scholar]
  432. Wan Ibrahim WN, Tofighi R, Onishchenko N, Rebellato P, Bose R, Uhlen P, Ceccatelli S. Perfluorooctane sulfonate induces neuronal and oligodendrocytic differentiation in neural stem cells and alters the expression of PPARgamma in vitro and in vivo. Toxicol Appl Pharmacol. 2013;269:51–60. doi: 10.1016/j.taap.2013.03.003. [DOI] [PubMed] [Google Scholar]
  433. Wang CY, Wang ZY, Xie JW, Cai JH, Wang T, Xu Y, Wang X, An L. CD36 upregulation mediated by intranasal LV-NRF2 treatment mitigates hypoxia-induced progression of Alzheimer’s-like pathogenesis. Antioxid Redox Signal. 2014;21:2208–2230. doi: 10.1089/ars.2014.5845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Wang G, Zhang J, Hu X, Zhang L, Mao L, Jiang X, Liou AK, Leak RK, Gao Y, Chen J. Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metab. 2013;33:1864–1874. doi: 10.1038/jcbfm.2013.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  435. Wang HE, Devereaux RS, Yealy DM, Safford MM, Howard G. National variation in United States sepsis mortality: a descriptive study. Int J Health Geogr. 2010;9:9. doi: 10.1186/1476-072X-9-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  436. Wang J, Rogove AD, Tsirka AE, Tsirka SE. Protective role of tuftsin fragment 1-3 in an animal model of intracerebral hemorrhage. Ann Neurol. 2003a;54:655–664. doi: 10.1002/ana.10750. [DOI] [PubMed] [Google Scholar]
  437. Wang J, Tsirka SE. Tuftsin fragment 1-3 is beneficial when delivered after the induction of intracerebral hemorrhage. Stroke. 2005;36:613–618. doi: 10.1161/01.STR.0000155729.12931.8f. [DOI] [PubMed] [Google Scholar]
  438. Wang N, Verna L, Chen NG, Chen J, Li H, Forman BM, Stemerman MB. Constitutive activation of peroxisome proliferator-activated receptor-gamma suppresses pro-inflammatory adhesion molecules in human vascular endothelial cells. J Biol Chem. 2002a;277:34176–34181. doi: 10.1074/jbc.M203436200. [DOI] [PubMed] [Google Scholar]
  439. Wang SH, Sun ZL, Guo YJ, Yuan Y, Li L. PPARgamma-mediated advanced glycation end products regulation of neural stem cells. Mol Cell Endocrinol. 2009;307:176–184. doi: 10.1016/j.mce.2009.02.012. [DOI] [PubMed] [Google Scholar]
  440. Wang X, Mao X, Xie L, Sun F, Greenberg DA, Jin K. Conditional depletion of neurogenesis inhibits long-term recovery after experimental stroke in mice. PLoS One. 2012a;7:e38932. doi: 10.1371/journal.pone.0038932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Wang Y, Huang J, Li Y, Yang GY. Roles of chemokine CXCL12 and its receptors in ischemic stroke. Curr Drug Targets. 2012b;13:166–172. doi: 10.2174/138945012799201603. [DOI] [PubMed] [Google Scholar]
  442. Wang Y, Liao J, Tang SJ, Shu J, Zhang W. HIV-1 gp120 Upregulates Brain-Derived Neurotrophic Factor (BDNF) Expression in BV2 Cells via the Wnt/beta-Catenin Signaling Pathway. J Mol Neurosci. 2017;62:199–208. doi: 10.1007/s12031-017-0931-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Wang Y, Liu G, Hong D, Chen F, Ji X, Cao G. White matter injury in ischemic stroke. Prog Neurobiol. 2016;141:45–60. doi: 10.1016/j.pneurobio.2016.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. Wang YL, Frauwirth KA, Rangwala SM, Lazar MA, Thompson CB. Thiazolidinedione activation of peroxisome proliferator-activated receptor gamma can enhance mitochondrial potential and promote cell survival. J Biol Chem. 2002b;277:31781–31788. doi: 10.1074/jbc.M204279200. [DOI] [PubMed] [Google Scholar]
  445. Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, Evans RM. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell. 2003b;113:159–170. doi: 10.1016/s0092-8674(03)00269-1. [DOI] [PubMed] [Google Scholar]
  446. Warden A, Truitt J, Merriman M, Ponomareva O, Jameson K, Ferguson LB, Mayfield RD, Harris RA. Localization of PPAR isotypes in the adult mouse and human brain. Sci Rep. 2016;6:27618. doi: 10.1038/srep27618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  447. Wardyn JD, Ponsford AH, Sanderson CM. Dissecting molecular cross-talk between Nrf2 and NF-kappaB response pathways. Biochem Soc Trans. 2015;43:621–626. doi: 10.1042/BST20150014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Wartenberg KE, Stoll A, Funk A, Meyer A, Schmidt JM, Berrouschot J. Infection after acute ischemic stroke: risk factors, biomarkers, and outcome. Stroke Res Treat. 2011;2011:830614. doi: 10.4061/2011/830614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Wasserman JK, Schlichter LC. White matter injury in young and aged rats after intracerebral hemorrhage. Exp Neurol. 2008;214:266–275. doi: 10.1016/j.expneurol.2008.08.010. [DOI] [PubMed] [Google Scholar]
  450. Wayman NS, Hattori Y, McDonald MC, Mota-Filipe H, Cuzzocrea S, Pisano B, Chatterjee PK, Thiemermann C. Ligands of the peroxisome proliferator-activated receptors (PPAR-gamma and PPAR-alpha) reduce myocardial infarct size. FASEB J. 2002;16:1027–1040. doi: 10.1096/fj.01-0793com. [DOI] [PubMed] [Google Scholar]
  451. Weiss N, Miller F, Cazaubon S, Couraud PO. The blood-brain barrier in brain homeostasis and neurological diseases. Biochim Biophys Acta. 2009;1788:842–857. doi: 10.1016/j.bbamem.2008.10.022. [DOI] [PubMed] [Google Scholar]
  452. Wilcox R, Bousser MG, Betteridge DJ, Schernthaner G, Pirags V, Kupfer S, Dormandy J, Investigators, P.R. Effects of pioglitazone in patients with type 2 diabetes with or without previous stroke: results from PROactive (PROspective pioglitAzone Clinical Trial In macroVascular Events 04) Stroke. 2007;38:865–873. doi: 10.1161/01.STR.0000257974.06317.49. [DOI] [PubMed] [Google Scholar]
  453. Willmot M, Leonardi-Bee J, Bath PM. High blood pressure in acute stroke and subsequent outcome: a systematic review. Hypertension. 2004;43:18–24. doi: 10.1161/01.HYP.0000105052.65787.35. [DOI] [PubMed] [Google Scholar]
  454. Wilson HM. Macrophages heterogeneity in atherosclerosis - implications for therapy. J Cell Mol Med. 2010;14:2055–2065. doi: 10.1111/j.1582-4934.2010.01121.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  455. Winkelmayer WC, Setoguchi S, Levin R, Solomon DH. Comparison of cardiovascular outcomes in elderly patients with diabetes who initiated rosiglitazone vs pioglitazone therapy. Arch Intern Med. 2008;168:2368–2375. doi: 10.1001/archinte.168.21.2368. [DOI] [PubMed] [Google Scholar]
  456. Wohlfert EA, Nichols FC, Nevius E, Clark RB. Peroxisome proliferator-activated receptor gamma (PPARgamma) and immunoregulation: enhancement of regulatory T cells through PPARgamma-dependent and -independent mechanisms. J Immunol. 2007;178:4129–4135. doi: 10.4049/jimmunol.178.7.4129. [DOI] [PubMed] [Google Scholar]
  457. Wolinski P, Glabinski A. Chemokines and neurodegeneration in the early stage of experimental ischemic stroke. Mediators Inflamm. 2013;2013:727189. doi: 10.1155/2013/727189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  458. Woo MS, Yang J, Beltran C, Cho S. Cell Surface CD36 Protein in Monocyte/Macrophage Contributes to Phagocytosis during the Resolution Phase of Ischemic Stroke in Mice. J Biol Chem. 2016;291:23654–23661. doi: 10.1074/jbc.M116.750018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  459. Writing Group, M. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB, American Heart Association Statistics, C. Stroke Statistics, S Heart Disease and Stroke Statistics-2016 Update: A Report From the American Heart Association. Circulation. 2016;133:e38–360. doi: 10.1161/CIR.0000000000000350. [DOI] [PubMed] [Google Scholar]
  460. Wu G, Wu J, Jiao Y, Wang L, Wang F, Zhang Y. Rosiglitazone infusion therapy following minimally invasive surgery for intracerebral hemorrhage evacuation decreases matrix metalloproteinase-9 and blood-brain barrier disruption in rabbits. BMC Neurol. 2015;15:37. doi: 10.1186/s12883-015-0287-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Wu JS, Cheung WM, Tsai YS, Chen YT, Fong WH, Tsai HD, Chen YC, Liou JY, Shyue SK, Chen JJ, Chen YE, Maeda N, Wu KK, Lin TN. Ligand-activated peroxisome proliferator-activated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation. 2009;119:1124–1134. doi: 10.1161/CIRCULATIONAHA.108.812537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  462. Wu JS, Tsai HD, Cheung WM, Hsu CY, Lin TN. PPAR-gamma Ameliorates Neuronal Apoptosis and Ischemic Brain Injury via Suppressing NF-kappaB-Driven p22phox Transcription. Mol Neurobiol. 2016;53:3626–3645. doi: 10.1007/s12035-015-9294-z. [DOI] [PubMed] [Google Scholar]
  463. Xiang Z, Lin T, Reeves SA. 15d-PGJ2 induces apoptosis of mouse oligodendrocyte precursor cells. J Neuroinflammation. 2007;4:18. doi: 10.1186/1742-2094-4-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  464. Xu R, Wang S, Li W, Liu Z, Tang J, Tang X. Activation of peroxisome proliferator-activated receptor-gamma by a 12/15-lipoxygenase product of arachidonic acid: a possible neuroprotective effect in the brain after experimental intracerebral hemorrhage. J Neurosurg. 2016:1–10. doi: 10.3171/2016.7.JNS1668. [DOI] [PubMed] [Google Scholar]
  465. Xu Y, Lu L, Greyson C, Lee J, Gen M, Kinugawa K, Long CS, Schwartz GG. Deleterious effects of acute treatment with a peroxisome proliferator-activated receptor-gamma activator in myocardial ischemia and reperfusion in pigs. Diabetes. 2003;52:1187–1194. doi: 10.2337/diabetes.52.5.1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Xu Y, Tamamaki N, Noda T, Kimura K, Itokazu Y, Matsumoto N, Dezawa M, Ide C. Neurogenesis in the ependymal layer of the adult rat 3rd ventricle. Exp Neurol. 2005;192:251–264. doi: 10.1016/j.expneurol.2004.12.021. [DOI] [PubMed] [Google Scholar]
  467. Xue M, Del Bigio MR. Intracerebral injection of autologous whole blood in rats: time course of inflammation and cell death. Neurosci Lett. 2000;283:230–232. doi: 10.1016/s0304-3940(00)00971-x. [DOI] [PubMed] [Google Scholar]
  468. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPARgamma/RXRalpha-induced and CD36- mediated microglial amyloid-beta phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 2012;32:17321–17331. doi: 10.1523/JNEUROSCI.1569-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Yamashita D, Yamaguchi T, Shimizu M, Nakata N, Hirose F, Osumi T. The transactivating function of peroxisome proliferator-activated receptor gamma is negatively regulated by SUMO conjugation in the amino-terminal domain. Genes Cells. 2004;9:1017–1029. doi: 10.1111/j.1365-2443.2004.00786.x. [DOI] [PubMed] [Google Scholar]
  470. Yang HC, Deleuze S, Zuo Y, Potthoff SA, Ma LJ, Fogo AB. The PPARgamma agonist pioglitazone ameliorates aging-related progressive renal injury. J Am Soc Nephrol. 2009a;20:2380–2388. doi: 10.1681/ASN.2008111138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  471. Yang J, Zhang D, Li J, Zhang X, Fan F, Guan Y. Role of PPARgamma in renoprotection in Type 2 diabetes: molecular mechanisms and therapeutic potential. Clin Sci (Lond) 2009b;116:17–26. doi: 10.1042/CS20070462. [DOI] [PubMed] [Google Scholar]
  472. Yang T, Michele DE, Park J, Smart AM, Lin Z, Brosius FC, 3rd, Schnermann JB, Briggs JP. Expression of peroxisomal proliferator-activated receptors and retinoid X receptors in the kidney. Am J Physiol. 1999;277:F966–973. doi: 10.1152/ajprenal.1999.277.6.F966. [DOI] [PubMed] [Google Scholar]
  473. Yang T, Sun Y, Lu Z, Leak RK, Zhang F. The impact of cerebrovascular aging on vascular cognitive impairment and dementia. Ageing Res Rev. 2017;34:15–29. doi: 10.1016/j.arr.2016.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  474. Yang T, Sun Y, Zhang F. Non-Neuronal Mechanisms of Brain Damage and Repair After Stroke. Springer; 2016. The Role of Nonneuronal Nrf2 Pathway in Ischemic Stroke: Damage Control and Potential Tissue Repair; pp. 377–397. [Google Scholar]
  475. Yao J, Zheng K, Zhang X. Rosiglitazone exerts neuroprotective effects via the suppression of neuronal autophagy and apoptosis in the cortex following traumatic brain injury. Mol Med Rep. 2015;12:6591–6597. doi: 10.3892/mmr.2015.4292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  476. Yessoufou A, Wahli W. Multifaceted roles of peroxisome proliferator-activated receptors (PPARs) at the cellular and whole organism levels. Swiss Med Wkly. 2010;140:w13071. doi: 10.4414/smw.2010.13071. [DOI] [PubMed] [Google Scholar]
  477. Yi JH, Park SW, Brooks N, Lang BT, Vemuganti R. PPARgamma agonist rosiglitazone is neuroprotective after traumatic brain injury via anti-inflammatory and anti-oxidative mechanisms. Brain Res. 2008;1244:164–172. doi: 10.1016/j.brainres.2008.09.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  478. Yilmaz G, Granger DN. Cell adhesion molecules and ischemic stroke. Neurol Res. 2008;30:783–793. doi: 10.1179/174313208X341085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  479. Yin KJ, Deng Z, Hamblin M, Xiang Y, Huang H, Zhang J, Jiang X, Wang Y, Chen YE. Peroxisome proliferator-activated receptor delta regulation of miR-15a in ischemia-induced cerebral vascular endothelial injury. J Neurosci. 2010;30:6398–6408. doi: 10.1523/JNEUROSCI.0780-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  480. Yin KJ, Fan Y, Hamblin M, Zhang J, Zhu T, Li S, Hawse JR, Subramaniam M, Song CZ, Urrutia R, Lin JD, Chen YE. KLF11 mediates PPARgamma cerebrovascular protection in ischaemic stroke. Brain. 2013;136:1274–1287. doi: 10.1093/brain/awt002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  481. Yin KJ, Hamblin M, Chen YE. Non-coding RNAs in cerebral endothelial pathophysiology: emerging roles in stroke. Neurochem Int. 2014;77:9–16. doi: 10.1016/j.neuint.2014.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  482. Yoo HY, Chang MS, Rho HM. Induction of the rat Cu/Zn superoxide dismutase gene through the peroxisome proliferator-responsive element by arachidonic acid. Gene. 1999;234:87–91. doi: 10.1016/s0378-1119(99)00176-6. [DOI] [PubMed] [Google Scholar]
  483. Yoshihara D, Kurahashi H, Morita M, Kugita M, Hiki Y, Aukema HM, Yamaguchi T, Calvet JP, Wallace DP, Nagao S. PPAR-gamma agonist ameliorates kidney and liver disease in an orthologous rat model of human autosomal recessive polycystic kidney disease. Am J Physiol Renal Physiol. 2011;300:F465–474. doi: 10.1152/ajprenal.00460.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Yoshii H, Onuma T, Yamazaki T, Watada H, Matsuhisa M, Matsumoto M, Kitagawa K, Kitakaze M, Yamasaki Y, Kawamori R, Group, P.J.S. Effects of pioglitazone on macrovascular events in patients with type 2 diabetes mellitus at high risk of stroke: the PROFIT-J study. J Atheroscler Thromb. 2014;21:563–573. [PubMed] [Google Scholar]
  485. Yu Y, Xue BJ, Wei SG, Zhang ZH, Beltz TG, Guo F, Johnson AK, Felder RB. Activation of central PPAR-gamma attenuates angiotensin II-induced hypertension. Hypertension. 2015;66:403–411. doi: 10.1161/HYPERTENSIONAHA.115.05726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Yue Tl TL, Chen J, Bao W, Narayanan PK, Bril A, Jiang W, Lysko PG, Gu JL, Boyce R, Zimmerman DM, Hart TK, Buckingham RE, Ohlstein EH. In vivo myocardial protection from ischemia/reperfusion injury by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation. 2001;104:2588–2594. doi: 10.1161/hc4601.099403. [DOI] [PubMed] [Google Scholar]
  487. Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn- SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med. 2002;33:337–349. doi: 10.1016/s0891-5849(02)00905-x. [DOI] [PubMed] [Google Scholar]
  488. Zhang M, An C, Gao Y, Leak RK, Chen J, Zhang F. Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection. Prog Neurobiol. 2013;100:30–47. doi: 10.1016/j.pneurobio.2012.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  489. Zhang Q, Hu W, Meng B, Tang T. PPARgamma agonist rosiglitazone is neuroprotective after traumatic spinal cord injury via anti-inflammatory in adult rats. Neurol Res. 2010;32:852–859. doi: 10.1179/016164110X12556180206112. [DOI] [PubMed] [Google Scholar]
  490. Zhang X, Rodriguez-Galan MC, Subleski JJ, Ortaldo JR, Hodge DL, Wang JM, Shimozato O, Reynolds DA, Young HA. Peroxisome proliferator-activated receptor-gamma and its ligands attenuate biologic functions of human natural killer cells. Blood. 2004a;104:3276–3284. doi: 10.1182/blood-2004-02-0664. [DOI] [PubMed] [Google Scholar]
  491. Zhang Y, Wang H, Li J, Jimenez DA, Levitan ES, Aizenman E, Rosenberg PA. Peroxynitrite-induced neuronal apoptosis is mediated by intracellular zinc release and 12-lipoxygenase activation. J Neurosci. 2004b;24:10616–10627. doi: 10.1523/JNEUROSCI.2469-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  492. Zhang Z, Chopp M. Vascular endothelial growth factor and angiopoietins in focal cerebral ischemia. Trends Cardiovasc Med. 2002;12:62–66. doi: 10.1016/s1050-1738(01)00149-9. [DOI] [PubMed] [Google Scholar]
  493. Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, Bruggen N, Chopp M. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106:829–838. doi: 10.1172/JCI9369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  494. Zhao X, Aronowski J. Immunological Mechanisms and Therapies in Brain Injuries and Stroke. Springer; 2014. The Role of PPARγ in Stroke; pp. 301–320. [Google Scholar]
  495. Zhao X, Grotta J, Gonzales N, Aronowski J. Hematoma resolution as a therapeutic target: the role of microglia/macrophages. Stroke. 2009a;40:S92–94. doi: 10.1161/STROKEAHA.108.533158. [DOI] [PubMed] [Google Scholar]
  496. Zhao X, Strong R, Zhang J, Sun G, Tsien JZ, Cui Z, Grotta JC, Aronowski J. Neuronal PPARgamma deficiency increases susceptibility to brain damage after cerebral ischemia. J Neurosci. 2009b;29:6186–6195. doi: 10.1523/JNEUROSCI.5857-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  497. Zhao X, Sun G, Ting SM, Song S, Zhang J, Edwards NJ, Aronowski J. Cleaning up after ICH: the role of Nrf2 in modulating microglia function and hematoma clearance. J Neurochem. 2015a;133:144–152. doi: 10.1111/jnc.12974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Zhao X, Sun G, Zhang J, Strong R, Song W, Gonzales N, Grotta JC, Aronowski J. Hematoma resolution as a target for intracerebral hemorrhage treatment: role for peroxisome proliferator-activated receptor gamma in microglia/macrophages. Ann Neurol. 2007;61:352–362. doi: 10.1002/ana.21097. [DOI] [PubMed] [Google Scholar]
  499. Zhao X, Zhang Y, Strong R, Grotta JC, Aronowski J. 15d-Prostaglandin J2 activates peroxisome proliferator-activated receptor-gamma, promotes expression of catalase, and reduces inflammation, behavioral dysfunction, and neuronal loss after intracerebral hemorrhage in rats. J Cereb Blood Flow Metab. 2006;26:811–820. doi: 10.1038/sj.jcbfm.9600233. [DOI] [PubMed] [Google Scholar]
  500. Zhao XR, Gonzales N, Aronowski J. Pleiotropic role of PPARgamma in intracerebral hemorrhage: an intricate system involving Nrf2, RXR, and NF-kappaB. CNS Neurosci Ther. 2015b;21:357–366. doi: 10.1111/cns.12350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  501. Zhao Z, Alam S, Oppenheim RW, Prevette DM, Evenson A, Parsadanian A. Overexpression of glial cell line-derived neurotrophic factor in the CNS rescues motoneurons from programmed cell death and promotes their long-term survival following axotomy. Exp Neurol. 2004;190:356–372. doi: 10.1016/j.expneurol.2004.06.015. [DOI] [PubMed] [Google Scholar]
  502. Zhou K, Zhong Q, Wang YC, Xiong XY, Meng ZY, Zhao T, Zhu WY, Liao MF, Wu LR, Yang YR, Liu J, Duan CM, Li J, Gong QW, Liu L, Yang MH, Xiong A, Wang J, Yang QW. Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL- 10/GSK3beta/PTEN axis. J Cereb Blood Flow Metab. 2017;37:967–979. doi: 10.1177/0271678X16648712. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  503. Zhu Y, Qi C, Korenberg JR, Chen XN, Noya D, Rao MS, Reddy JK. Structural organization of mouse peroxisome proliferator-activated receptor gamma (mPPAR gamma) gene: alternative promoter use and different splicing yield two mPPAR gamma isoforms. Proc Natl Acad Sci U S A. 1995;92:7921–7925. doi: 10.1073/pnas.92.17.7921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  504. Zoete V, Grosdidier A, Michielin O. Peroxisome proliferator-activated receptor structures: ligand specificity, molecular switch and interactions with regulators. Biochim Biophys Acta. 2007;1771:915–925. doi: 10.1016/j.bbalip.2007.01.007. [DOI] [PubMed] [Google Scholar]
  505. Zuhayra M, Zhao Y, von Forstner C, Henze E, Gohlke P, Culman J, Lutzen U. Activation of cerebral peroxisome proliferator-activated receptors gamma (PPARgamma) reduces neuronal damage in the substantia nigra after transient focal cerebral ischaemia in the rat. Neuropathol Appl Neurobiol. 2011;37:738–752. doi: 10.1111/j.1365-2990.2011.01169.x. [DOI] [PubMed] [Google Scholar]
  506. Zuo Y, Yang HC, Potthoff SA, Najafian B, Kon V, Ma LJ, Fogo AB. Protective effects of PPARgamma agonist in acute nephrotic syndrome. Nephrol Dial Transplant. 2012;27:174–181. doi: 10.1093/ndt/gfr240. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES