Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2010 Sep 23;3(4):283–292.

Peroxisome proliferator activated receptor-γ and traumatic brain injury

Lei Qi 1,2,3, Asha Jacob 1,2, Ping Wang 1,2, Rongqian Wu 1,2
PMCID: PMC2971540  PMID: 21072262

Abstract

Traumatic brain injury (TBI) represents a major health care problem and a significant socioeconomic challenge worldwide. No specific therapy for TBI is available. The peroxisome proliferator activated receptor-γ (PPAR-γ) belongs to the nuclear receptor superfamily. Although PPAR-γ was originally characterized in adipose tissue as a regulator of lipid and glucose metabolism, recent studies showed that PPAR-γ is present in most cell types and plays a central role in the regulation of adipogenesis, glucose homeostasis, cellular differentiation, apoptosis and inflammation. Here, we reviewed the current literature on the molecular mechanisms of PPAR-γ-related neuroprotection after TBI. Growing evidence has indicated that the beneficial effects of PPAR-γ activation in TBI appear to be mediated through downregulation of inflammatory responses, reduction of oxidative stress, inhibition of apoptosis, and promotion of neurogenesis. A thorough understanding of the PPAR-γ pathway will be critical to the development of therapeutic interventions for the treatment of patients with TBI.

Keywords: Peroxisome proliferator activated receptor-γ (PPAR-γ), traumatic brain injury (TBI), nuclear receptor super-family, neuroprotection, neurogenesis, oxidative stress

Introduction

Traumatic brain injury (TBI) represents a major health care problem and a significant socioeconomic challenge worldwide [1-4]. Despite advances in research and improved neurological intensive care in recent years, the clinical outcome of severely head-injured patients is still poor. Survivors of TBI are often left with significant cognitive, behavioral, and communicative disabilities, and some patients develop long-term medical complications [4,5]. The annual economic burden of direct and indirect costs for TBI in the United States alone is estimated at more than $50 billion [6,7]. In its ‘World report on traffic injury prevention', the World Health Organization estimates that by 2020, road traffic accidents, which often cause head injury, would be within the top three leading causes of global burden of diseases, surpassing even HIV and tuberculosis. These statistics underline the urgent need for efficient treatment modalities to improve post-traumatic morbidity and mortality.

The damage to the brain after head trauma occurs in two phases: the initial primary phase and an ongoing secondary phase. The initial phase is the injury itself (i.e., immediate mechanical disruption of brain tissue). It is irreversible and amenable only to preventive measures. The secondary phase begins at the time of injury and continues in the ensuing days to weeks. This delayed phase leads to a variety of physiological, cellular, and molecular responses aimed at restoring the homeostasis of the damaged tissue, which, if not controlled, will intensify the damage sustained following TBI. Understanding of the cellular and molecular mechanisms contributing to the development of secondary brain injury after TBI is essential for the identification of new potential therapeutic targets.

Peroxisome proliferator activated receptors (PPARs) are members of the nuclear receptor superfamily. They can heterodimerize with reti-noid X receptor to regulate transcription of genes linked to lipid and glucose metabolism. In 1987, a peroxisome proliferator-binding protein was identified and characterized in the rat liver [8]. The first PPAR was subsequently cloned from the mouse liver [9], followed by other PPAR homologs in other species [10]. Three different PPARs [a,δ (also called β), γ] have been identified [11]. PPAR-γ is the most extensively studied of the three PPAR subtypes to date. PPAR-γ is remarkably conservative across species. Human and murine PPAR-γ proteins show 95% identity at the amino acid level. Human PPAR-γ gene is located on chromosome 3 whereas mouse PPAR-γ gene is located on chromosome 6. Mouse and human PPAR-γ genes reveal a common organization of the translated region in six exons. The genes extend over 100 kb of genomic DNA and generate three mRNA transcripts, PPAR-γ1, PPAR-γ2 and PPAR-γ3, which arise as products of different promoter usage and splicing [12-14]. PPAR-γ1 and PPAR-γ3 mRNAs encode for the same protein, while PPAR-γ2 mRNA gives rise to a protein containing 28 additional amino acids at the N-terminus. Analysis of the tissue distribution of the three mRNA PPAR-γ isoforms revealed some differences. PPAR-γ1 has the broadest tissue expression, while PPAR-γ2 and PPAR-γ3 isoforms are more restrictedly distributed.

PPAR-γ was originally identified as a key regulator of adipocyte differentiation and lipid metabolism [15] and the role of PPAR-γ in regulating glucose homeostasis has also been established [16]. It has been demonstrated that the anti-diabetic drugs known as thiazolidinediones (i.e., rosiglitazone, pioglitazone, troglitazone and cigli-tazone) mediate their therapeutic effects through the interaction with PPAR-γ [17]. In addition, several endogenous ligands of PPAR-γ have been identified, including prostaglandin derivatives such as 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) [18]. Studies from our laboratory have shown that some non-PPAR-y ligands such as vasoactive hormone adrenomedullin and its bind protein complex (AM/AMBP-1) [19] and dietary polyphenol cur-cumin [20,21] can also activate the PPAR-γ pathway. PPAR-γ gene silencing eliminated their beneficial effects ([19] and unpublished data). Recently, a few specific PPAR-γ antagonists were also identified. GW9662, a potent irreversible PPAR-γ ligand, is able to block PPAR-Y at 1-10 μM concentrations [22]. LG100641, another PPAR-γ ligand, does not activate PPAR-γ but selectively and competitively blocks thiazolidinediones' activation of the receptor [23]. Although PPAR-g has been investigated for effects of its ligands in modulating lipid metabolism, more and more recent studies have been showing the central role of PPAR-γ in the regulation of adipogenesis [24], glucose homeostasis [25], cellular differentiation [26], apoptosis [27] and inflammation [28,29]. Here, we present a review of the current literature on the molecular mechanisms of PPAR-γ-related neuroprotection after TBI.

PPAR-γand cerebral inflammation

TBI initiates an acute inflammatory response, which involves the integrated activities of cytokines, chemokines, vascular adhesion molecules, and inflammatory cells [30]. The traumatic injury to brain tissue gives rise to immediate mechanical damage characterized by disruption of cell membranes, cellular organelles, and blood vessels, which causes primary cell death, ischemia, and hemorrhage. These initial events then stimulate resident inflammatory cells in central nervous system (CNS) such as microglia and astrocytes and trigger the release of inflammatory mediators including cytokines, chemokines, neurotransmitters and reactive oxygen radicals or species, which in turn disrupt the blood brain barrier (BBB) and recruit more inflammatory cells [30-32]. These cells then release more inflammatory mediators and exaggerate the inflammatory response. This inflammatory cascade contributes to the acute pathologic processes and long-term neuronal damage following TBI [33].

PPAR-γ plays a major role in the regulation of inflammatory responses [29]. Increasing evidence has shown that PPAR-γ activation mitigates inflammation associated with chronic and acute neurological insults [34-37]. Activation of PPAR-γ in microglia and macrophages can decrease the production of proinflammatory mediators [38,39]. In fact, PPAR-γ induction in microglia and macrophages correlates with decreased microglia l and macrophage activation, decreased release of pro-inflammatory mediators and improved neurological outcome [40,41]. Thiazolidinediones have been shown to be beneficial in several cellular and animal models of CNS diseases where inflammation is a major component of the progressive neurological deficit [41-44]. Interestingly, a recent study has shown that cortical PPAR-γ expression is upregulated after TBI, and administration of PPAR-γ agonists is protective under such a condition [45]. Using a mouse model of TBI induced by controlled cortical impact, Yi etal. [45] showed that PPAR-γ mRNA expression in the cortical tissue surrounding the injury epicenter was more than doubled at 24 h after TBI. Administration of rosiglitazone, a PPAR-γ agonist, further increased PPAR-γ expression, reduced GSI-B4 (a marker of activated microglia/macrophages) and intercellular adhesion molecule-1 (ICAM-1) immunostaining, and downregulated pro-inflammatory cytokine expression in the brain after TBI. Similarly, Hyong et al. [46] showed that rosiglitazone significantly attenuated myeloperoxidase (MPO) activity (a measure of neutrophil infiltration) and decreased tumor necrosis factor-a (TNF-a) and interleukin (IL)-1β expression in a rat model of surgical brain injury.

Several pathways have been associated with PPAR-γ-dependent anti-inflammation [29,47, 48]. PPAR-γ can block nuclear factor (NF)-κB-dependent gene expression. Although nuclear receptors repress target genes in the absence of ligands by recruiting corepressors, the molecular mechanism for transcriptional repression by nuclear receptors in response to the binding of ligands remains poorly understood. One possibility involves the reciprocal inhibition of differential transcription systems through limited availability of shared cofactors, sometimes referred to as squelching [29]. PPAR-γ can scavenge transcriptional co-factors, such as steroid receptor co-activator-1 (SRC-1) and cAMP response element-binding protein (CREB) binding protein (CBP/p300). Bound to PPAR-γ, these co-factors are not available for initiating gene expression. Because transcriptional co-factors are indispensable for NF-κB-dependent gene induction, NF-κB-mediated gene expression is down regulated. NF-κB is an important transcription factor involved in pro-inflammatory gene expression. Blocking it can cause a significant attenuation of pro-inflammatory gene expression. A recent report by Glass and colleagues [49] suggests an alternative mechanism, wherein a functionally distinct pool of PPAR-γ is susceptible to ligand-dependentsumoylation at lysine 365, leading to recruitment to and stabilization of the nuclear receptor co-repressor (NCo-R)-histone deacety-lase-3 (HDAC3)-transducin beta-like protein 1 (TBL1)-TBLR1 complex at the promoters of pro-inflammatory genes. Thereby, NF-κB-dependent pro-inflammatory gene expression is abolished. Besides inactivating NF-κB, PPAR-γ can also directly associate with other transcription factors such as nuclear factor of activated T cells (NF-AT), activator protein-1 (AP-1), or transducers and activators of transcription (STAT), and block these transcription factors-dependent gene expression [29]. Additionally, PPAR-γ can inhibit phosphorylation of mitogen-activated protein kinase (MAPK), and therefore prevent MAPK-dependent pro-inflammatory gene expression [50].

PPAR-γ and oxidative stress

Oxidative damage is a significant component of the secondary injury cascade following TBI. A massive production of reactive oxygen species is triggered by increased intracellular calcium levels, mitochondrial dysfunction, arachidonic acid breakdown, activation of cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS), and glutamate-mediated excitotoxicity after brain trauma [51,52]. The oxidative cascade begins with the production of superoxide and nitric oxide. These two molecules combine to form peroxynitrite, which leads to the production of multiple damaging free radicals, including nitrogen dioxide, the carbonate radical, and the hydroxyl radical. Together, these free radicals lead to oxidative damage and secondary brain injury [53].

PPAR-γ activation affects the generation of reactive oxygen species on various levels. Systemic and intracerebroventricular administration of thiazolidinedione PPAR-γ agonists reduced the expression of COX-2, an important enzyme involved in the production of reactive oxygen species [54,55]. PPAR-γ agonists also attenuated the expression of iNOS in inflammatory cells [56,57], which is considered to be an important source of the deleterious radical peroxynitrite. On the other hand, PPAR-γ agonists also increase the levels of antioxidants in and around the injured tissue. Pioglitazone has been shown to induce the expression of the antioxidant enzyme superoxide dismutase, which scavenges free oxygen radicals in the injured tissue [58].

Elevated catalase levels were observed in PPAR-γ agonist-treated animals following intracerebral hemorrhage [59]. A recent study has also shown that PPAR-γ agonists reversed the depleted stores of glutathione in the hippocampus in a rat model of stroke [55]. In this regard, PPAR-γ activation may reduce the production of free radicals, and at the same time, elevate enzymes essential for combating free radicals that remain.

PPAR-γ and neural apoptosis

Neuronal apoptosis occurs in naturally physiologic states such as CNS development, as well as pathologic states such as ischemia, viral infection, neurodegenerative diseases, and trauma [60]. Both animal and human studies have shown substantial evidence of neuronal apoptosis after TBI [61]. After a brain insult, the injured tissue usually suffers from two waves of cell death, presumably representing two waves of neuronal death, the first of which is necrotic cell death and the second of which represents a delayed neuronal death by apoptosis. In rat models of TBI, necrosis was suggested to occur in the early post-traumatic period and apoptosis was observed in the chronic post-traumatic period [62]. Decreases in bcl-xl, an inhibitor of apoptosis, and increases in BAX, a promoter of apoptosis, were shown in the rat brain after TBI [63]. TUN EL positive cells were increased after experimental TBI [64,65]. Activation of caspases 3, 7, 9, and 12 was reported in a variety of animal models of TBI [66-68]. In humans, swollen hippocampal neurons in contusions were observed in the early post-traumatic period. These injured neurons eventually shrink and become eosinophilic over time. TUNEL positive cells were observed in histologic samples of TBI patients [69,70]. Caspase 8 was upregulated in the human brain after TBI [71]. Clearly, therapies that protect surrounding cells of the necrotic core, the so-called penumbra, from apoptosis are of great importance, because the loss of these cells contributes to the loss of neurological function and can greatly impact outcome.

PPAR-γ agonists have been shown to increase neuron survival and decrease lesion sizes in animal models of Parkinson's disease [72,73], central inflammation [74], intracerebral hemorrhage [59], and cerebral ischemia [75,76]. In experimental models of TBI, PPAR-γ agonists have also been shown to inhibit neuronal apoptosis. Yi et al. [45] showed that the cortical lesion volume was significantly decreased by rosiglitazone treatment, which was associated with less numbers of TUNEL positive neurons and curtailed induction of caspase 3 and Bax after TBI. These beneficial effects were likely mediated through an indirect mechanism involving the anti-inflammatory and anti-oxidative effects of PPAR-γ. However, PPAR-γ activation may also have a direct anti-apoptotic effect on neurons. Neuronal expression of PPAR-γ has been detected in the intact CNS and an upregu-lation of PPAR-γ was observed in neurons in the ischemic penumbra following focal cerebral ischemia [75,76]. Using both in vitro and in vivo models of excitotoxic neuronal injury, Zhao etal. [77] have shown that the endogenous PPAR-γ ligand, 15d-PGJ2, and a selective thiazolidinedione PPAR-γ agonist, ciglitazone, significantly reduced glutamate and NMDA-mediated neuronal death. This neuroprotective effect of 15d-PGJ2 and ciglitazone was linked to increased PPAR-γ DNA binding activity as it was fully reversed by the pretreatment of neurons with selective PPAR-γ antagonists and anti-PPAR-γ antibody.

Interestingly, PPAR-γ agonists have been shown to induce apoptosis in glioma and neuroblastoma cells. In four different glioma cell lines (A172, U87-MG, M059K, M059J), rosiglitazone led to the induction of apoptosis and the PPAR-γ antagonist GW9662 partially reverted these effects [78]. Ciglitazone and 15d-PGJ2 also induced apoptotic cell death in human and rat glioma cells, and apoptotic cell death was correlated with the upregulation of Bax and Bad protein levels [79,80]. In cultured human neuro-blastoma cells, 15d-PGJ2 was shown to decrease cellular viability and induce apoptosis [81,82]. The precise mechanisms responsible for differential effects (i.e. pro-apoptotic vs. anti-apoptotic) of PPAR-γ ligands remain incompletely clarified. Certainly, indirect mechanisms such as anti-inflammation and anti-oxidation contribute to the anti-apoptotic effect of PPAR-γ agonists observed in the above mentioned animal models. However, the different effects of PPAR-γ agonists on apoptosis may also be related with the concentrations of PPAR-γ agonists used [83]. Clay etal. [84] have pointed out that modest activation of PPAR-γ results in enhancement of cellular proliferation, whereas vigorous activation might induce apoptosis. Whether PPAR-γ ligands induce apoptosis or stimulate cell proliferation may depend on the direction of propagation of intracellular signals involved, which reflects the concentration-dependent effect of PPAR-γ ligands on PPAR-γ activation.

PPAR-γ and neurogenesis

During development and throughout our lifetime neuronal death, maintenance and neurogenesis occur in a tightly controlled fashion [85,86]. The neurogenic zones, replete with neural stem cells are found in the subventricular zones, located along the lateral aspects of the lateral ventricles and the subgranular zones, located in the dentate gyrus of the hippocampus. Adult neurogenesis is found in these forebrain regions in all mammalian species examined, including humans [87,88], and may serve to replace cells damaged by brain insults. Normally, the postnatal subventricular zones contributes progenitors to the rostral migratory stream to support ongoing olfactory neurogenesis, while the subgranular zones of the dentate gyrus provides new granular neurons throughout life [89,90]. Following TBI, neural stem cells, supported by their local vasculature, are thought to proliferate, migrate to and differentiate at injury sites, affecting variable degrees of structural and functional recovery [91]. Significant self-recovery occurs following all but the most severe episodes of TBI [92-94]. Although the mechanisms underlying this phenomenon remain largely unknown, it is clear that neural stem cells are activated and increased neurogenesis follows. New neurons are found in the outer layers of the dentate gyrus, where normally this would occur only during early development [95-97]. Injury-induced neurogenesis is one compelling potential contributor to post-injury recovery [91].

PPAR-γ appears to be important in regulating the early brain development and post-injury brain repair [98]. PPAR-γ expression is relatively low in newborn and adult brains. But high levels of PPAR-γ expression were found in embryonic brains [99]. Similarly, neural stem cells isolated from mouse embryos also express high levels of PPAR-γ. Compared with wild-type mice, neural stem cells isolated from heterozygous PPAR-γ-deficient mice exhibited a slower growth rate [100]. Suppression of PPAR-γ protein expression by a lentivirus-mediated short hairpin RNA technique led to a significant reduction in the cell growth rate in neural stem cells [98]. In the case of control infection, which was unable to suppress the expression of PPAR-γ protein, the proliferation of neural stem cells was not altered. Similarly, optimal activation of the PPAR-γ pathway by PPAR-γ agonists stimulated neural stem cell proliferation and inhibited differentiation of neural stem cells into neurons. On the other hand, PPAR-γ antagonists inhibited cell growth and induced apoptosis/death in neural stem cells [100].

Similar to PPAR-γ's effect on apoptosis, PPAR-γ's regulation of neural stem cells is also biphasic. Physiological concentrations of PPAR-γ stimulated neural stem cell growth, whereas excessive activation of PPAR-γ with higher concentrations of agonists resulted in cell death [100]. A recent study has shown that PPAR-γ agonists at concentrations of 100 nM to 3 μM stimulated neural stem cell growth. However, inhibition of cell growth and apoptosis were conversely observed when PPAR-γ agonists' concentrations were above 30 μM [100]. This biphasic effect of PPAR-γ suggests that optimal concentrations of PPAR-γ agonists exist for neurogenesis.

Brain repair/recovery after injury involves both angiogenesis and neurogenesis. PPAR-γ agonists have also been shown to promote angiogenesis in the brain by their action on endothe-lial progenitor cells [101]. In a rat model of focal cerebral ischemia, Chu et al. [102] have found that rosiglitazone treatment increased the vascular surface area, the vascular branch points, the vascular length, and the number of BrdU (5-bromo-2-deoxyuridine, a marker for dividing cells) positive endothelial cells. Thus, PPAR-γ agonists may also influence brain repair through angiogenesis.

Summary and perspectives

TBI is a major disabling condition all over the world. No specific pharmacological therapy for TBI is available to reduce the incidence of secondary brain injury and adverse outcome. Several pathological events including inflammation, oxidative stress and apoptosis during the acute stage after the injury are known to precipitate the neuronal death and neurological dysfunction. In this review, we highlighted the beneficial effects of PPAR-γ agonists in TBI. The potential molecular mechanisms of PPAR-γ action were also discussed. The beneficial effects of PPAR-γ agonists in TBI appear to be mediated through the downregulation of inflammatory responses, reduction of oxidative stress, inhibition of apoptosis, and promotion of neurogene-sis. However, the exact role of PPAR-γ following TBI remains unclear. And our knowledge of the regulatory mechanisms and signaling cascades underlying the beneficial effects of PPAR-γ in the brain is still limited. Further research in these areas is required. Nevertheless, there is increasing evidence that a thorough understanding of the PPAR-γ pathway will be critical to the development of therapeutic interventions for the treatment of TBI patients.

Acknowledgments

This work was supported by a DoD/DRMRP Grant DR080669 (to R.Wu).

References

  • 1.Aarabi B, Simard JM. Traumatic brain injury. Curr Opin Crit Care. 2009;15:548–553. doi: 10.1097/MCC.0b013e32833190da. [DOI] [PubMed] [Google Scholar]
  • 2.Marmarou A, Lu J, Butcher I, McHugh GS, Mushkudiani NA, Murray GD, Steyerberg EW, Maas AI. IMPACT database of traumatic brain injury: design and description. J Neurotrauma. 2007;24:239–250. doi: 10.1089/neu.2006.0036. [DOI] [PubMed] [Google Scholar]
  • 3.Margulies S, Hicks R. Combination therapies for traumatic brain injury: prospective considerations. J Neurotrauma. 2009;26:925–939. doi: 10.1089/neu.2008.0794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Maas AI, Marmarou A, Murray GD, Teasdale SG, Steyerberg EW. Prognosis and clinical trial design in traumatic brain injury: the IMPACT study. J Neurotrauma. 2007;24:232–238. doi: 10.1089/neu.2006.0024. [DOI] [PubMed] [Google Scholar]
  • 5.Bales JW, Wagner AK, Kline AE, Dixon CE. Persistent cognitive dysfunction after traumatic brain injury: A dopamine hypothesis. Neurosci Biobehav Rev. 2009;33:981–1003. doi: 10.1016/j.neubiorev.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Davis KL, Joshi AV, Tortella BJ, Candrilli SD. The direct economic burden of blunt and penetrating trauma in a managed care population. J Trauma. 2007;62:622–629. doi: 10.1097/TA.0b013e318031afe3. [DOI] [PubMed] [Google Scholar]
  • 7.Boulanger L, Joshi AV, Tortella BJ, Menzin J, Caloyeras JP, Russell MW. Excess mortality, length of stay, and costs associated with serious hemorrhage among trauma patients: findings from the National Trauma Data Bank. Am Surg. 2007;73:1269–1274. [PubMed] [Google Scholar]
  • 8.Lalwani ND, Alvares K, Reddy MK, Reddy MN, Parikh I, Reddy JK. Peroxisome proliferator-binding proteidentification and partial characterization of nafenopin-, clofibric acid-, and ciprofibrate-binding proteins from rat liver. Proc Natl Acad Sci U S A. 1987;84:5242–5246. doi: 10.1073/pnas.84.15.5242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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]
  • 10.Schoonjans K, Staels B, Auwerx J. Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression. J Lipid Res. 1996;37:907–925. [PubMed] [Google Scholar]
  • 11.Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–839. doi: 10.1016/0092-8674(95)90199-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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]
  • 13.Hotta K, Gustafson TA, Yoshioka S, Ortmeyer HK, Bodkin NL, Hansen BC. Relationships of PPARgamma and PPARgamma2 mRNA levels to obesity, diabetes and hyperinsulinaemia in rhesus monkeys. Int J Obes Relat Metab Disord. 1998;22:1000–1010. doi: 10.1038/sj.ijo.0800718. [DOI] [PubMed] [Google Scholar]
  • 14.Fajas L, Fruchart JC, Auwerx J. PPARgamma3 mRNA: a distinct PPARgamma mRNA subtype transcribed from an independent promoter. FEBS Lett. 1998;438:55–60. doi: 10.1016/s0014-5793(98)01273-3. [DOI] [PubMed] [Google Scholar]
  • 15.Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79:1147–1156. doi: 10.1016/0092-8674(94)90006-x. [DOI] [PubMed] [Google Scholar]
  • 16.Chinetti G, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm Res. 2000;49:497–505. doi: 10.1007/s000110050622. [DOI] [PubMed] [Google Scholar]
  • 17.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]
  • 18.Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem. 1997;272:3406–3410. doi: 10.1074/jbc.272.6.3406. [DOI] [PubMed] [Google Scholar]
  • 19.Miksa M, Wu R, Cui X, Dong W, Das P, Simms HH, Ravikumar TS, Wang P. Vasoactive hormone adrenomedullin and its binding proteanti-inflammatory effects by up-regulating peroxisome proliferator-activated receptor-gamma. J Immunol. 2007;179:6263–6272. doi: 10.4049/jimmunol.179.9.6263. [DOI] [PubMed] [Google Scholar]
  • 20.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]
  • 21.Jacob A, Wu R, Zhou M, Wang P. Mechanism of the Anti-inflammatory Effect of CurcumPPAR-gamma Activation. PPAR Res. 2007;2007:89369. doi: 10.1155/2007/89369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perez-Ortiz JM, Tranque P, Vaquero CF, Domingo B, Molina F, Calvo S, Jordan J, Cena V, Llopis J. Glitazones differentially regulate primary astro-cyte and glioma cell survival. Involvement of reactive oxygen species and peroxisome proliferator-activated receptor-gamma. J Biol Chem. 2004;279:8976–8985. doi: 10.1074/jbc.M308518200. [DOI] [PubMed] [Google Scholar]
  • 23.Mukherjee R, Hoener PA, Jow L, Bilakovics J, Klausing K, Mais DE, Faulkner A, Croston GE, Paterniti JR., Jr A selective peroxisome proliferator-activated receptor-gamma (PPARgamma) modulator blocks adipocyte differentiation but stimulates glucose uptake in 3T3-L1 adipocytes. Mol Endocrinol. 2000;14:1425–1433. doi: 10.1210/mend.14.9.0528. [DOI] [PubMed] [Google Scholar]
  • 24.Willson TM, Lambert MH, Kliewer SA. Peroxisome proliferator-activated receptor gamma and metabolic disease. Annu Rev Biochem. 2001;70:341–367. doi: 10.1146/annurev.biochem.70.1.341. [DOI] [PubMed] [Google Scholar]
  • 25.Nolan JJ, Ludvik B, Beerdsen P, Joyce M, Olefsky J. Improvement in glucose tolerance and insulin resistance in obese subjects treated with troglitazone. N Engl J Med. 1994;331:1188–1193. doi: 10.1056/NEJM199411033311803. [DOI] [PubMed] [Google Scholar]
  • 26.Tontonoz P, Singer S, Forman BM, Sarraf P, Fletcher JA, Fletcher CD, Brun RP, Mueller E, Altiok S, Oppenheim H, Evans RM, Spiegelman BM. Terminal differentiation of human liposarcoma cells induced by ligands for peroxisome proliferator-activated receptor gamma and the retinoid X receptor. Proc Natl Acad Sci U S A. 1997;94:237–241. doi: 10.1073/pnas.94.1.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chang TH, Szabo E. Induction of differentiation and apoptosis by ligands of peroxisome proliferator-activated receptor gamma in non-small cell lung cancer. Cancer Res. 2000;60:1129–1138. [PubMed] [Google Scholar]
  • 28.Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response. J Clin Invest. 1999;104:383–389. doi: 10.1172/JCI7145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lehrke M, Lazar MA. The many faces of PPARgamma. Cell. 2005;123:993–999. doi: 10.1016/j.cell.2005.11.026. [DOI] [PubMed] [Google Scholar]
  • 30.Cederberg D, Siesjo P. What has inflammation to do with traumatic brain injury? Childs Nerv Syst. 2010;26:221–226. doi: 10.1007/s00381-009-1029-x. [DOI] [PubMed] [Google Scholar]
  • 31.Yong VW, Rivest S. Taking advantage of the systemic immune system to cure brain diseases. Neuron. 2009;64:55–60. doi: 10.1016/j.neuron.2009.09.035. [DOI] [PubMed] [Google Scholar]
  • 32.Lu J, Goh SJ, Tng PY, Deng YY, Ling EA, Moochhala S. Systemic inflammatory response following acute traumatic brain injury. Front Biosci. 2009;14:3795–3813. doi: 10.2741/3489. [DOI] [PubMed] [Google Scholar]
  • 33.Ziebell JM, Morganti-Kossmann MC. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics. 2010;7:22–30. doi: 10.1016/j.nurt.2009.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sundararajan S, Jiang Q, Heneka M, Lan-dreth G. PPARgamma as a therapeutic target in central nervous system diseases. Neurochem Int. 2006;49:136–144. doi: 10.1016/j.neuint.2006.03.020. [DOI] [PubMed] [Google Scholar]
  • 35.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]
  • 36.Bernardo A, Minghetti L. PPAR-gamma agonists as regulators of microglial activation and brain inflammation. Curr Pharm Des. 2006;12:93–109. doi: 10.2174/138161206780574579. [DOI] [PubMed] [Google Scholar]
  • 37.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]
  • 38.Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macro-phage activation. Nature. 1998;391:79–82. doi: 10.1038/34178. [DOI] [PubMed] [Google Scholar]
  • 39.Petrova TV, Akama KT, Van Eldik LJ. Cyclopentenone prostaglandins suppress activation of microglia: down-regulation of inducible nitric-oxide synthase by 15-deoxy-Delta12,14-prostaglandin J2. Proc Natl Acad Sci USA. 1999;96:4668–4673. doi: 10.1073/pnas.96.8.4668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Steinman L, Martin R, Bernard C, Conlon P, Oksenberg JR. Multiple sclerosis: deeper understanding of its pathogenesis reveals new targets for therapy. Annu Rev Neurosci. 2002;25:491–505. doi: 10.1146/annurev.neuro.25.112701.142913. [DOI] [PubMed] [Google Scholar]
  • 41.Mrak RE, Landreth GE. PPARgamma, neuroin-flammation, and disease. J Neuroinflammation. 2004;1:5. doi: 10.1186/1742-2094-1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Diab A, Deng C, Smith JD, Hussain RZ, Phanavanh B, Lovett-Racke AE, Drew PD, Racke MK. Peroxisome proliferator-activated receptor-gamma agonist 15-deoxy-Delta(12,14)-prostaglandin J(2) ameliorates experimental autoimmune encephalomyelitis. J Immunol. 2002;168:2508–2515. doi: 10.4049/jimmunol.168.5.2508. [DOI] [PubMed] [Google Scholar]
  • 43.Kiaei M, Kipiani K, Chen J, Calingasan NY, Beal MF. Peroxisome proliferator-activated receptor-gamma agonist extends survival in transgenic mouse model of amyotrophic lateral sclerosis. Exp Neurol. 2005;191:331–336. doi: 10.1016/j.expneurol.2004.10.007. [DOI] [PubMed] [Google Scholar]
  • 44.Landreth GE, Heneka MT. Anti-inflammatory actions of peroxisome proliferator-activated receptor gamma agonists in Alzheimer's disease. Neurobiol Aging. 2001;22:937–944. doi: 10.1016/s0197-4580(01)00296-2. [DOI] [PubMed] [Google Scholar]
  • 45.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]
  • 46.Hyong A, Jadhav V, Lee S, Tong W, Rowe J, Zhang JH, Tang J. Rosiglitazone, a PPAR gamma agonist, attenuates inflammation after surgical brain injury in rodents. Brain Res. 2008;1215:218–224. doi: 10.1016/j.brainres.2008.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.von KA, Soller M, Brune B. Peroxisome proliferator-activated receptor gamma (PPAR gamma) and sepsis. Arch Immunol Ther Exp (Warsz ) 2007;55:19–25. doi: 10.1007/s00005-007-0005-y. [DOI] [PubMed] [Google Scholar]
  • 48.Szanto A, Nagy L. The many faces of PPARgamma: anti-inflammatory by any means? Immunobiology. 2008;213:789–803. doi: 10.1016/j.imbio.2008.07.015. [DOI] [PubMed] [Google Scholar]
  • 49.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]
  • 50.Desreumaux P, Dubuquoy L, Nutten S, Peuch-maur M, Englaro W, Schoonjans K, Derijard B, Desvergne B, Wahli W, Chambon P, Leibowitz MD, Colombel JF, Auwerx J. Attenuation of colon inflammation through activators of the retinoid X receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma) heterodimer. A basis for new therapeutic strategies. J Exp Med. 2001;193:827–838. doi: 10.1084/jem.193.7.827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bayir H, Kochanek PM, Kagan VE. Oxidative stress in immature brain after traumatic brain injury. Dev Neurosci. 2006;28:420–431. doi: 10.1159/000094168. [DOI] [PubMed] [Google Scholar]
  • 52.Eghwrudjakpor PO, Allison AB. Oxidative stress following traumatic brain injury: enhancement of endogenous antioxidant defense systems and the promise of improved outcome. Niger J Med. 2010;19:14–21. doi: 10.4314/njm.v19i1.52466. [DOI] [PubMed] [Google Scholar]
  • 53.Potts MB, Koh SE, Whetstone WD, Walker BA, Yoneyama T, Claus CP, Manvelyan HM, Noble-Haeusslein LJ. Traumatic injury to the immature brainflammation, oxidative injury, and iron-mediated damage as potential therapeutic targets. NeuroRx. 2006;3:143–153. doi: 10.1016/j.nurx.2006.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhao Y, Patzer A, Herdegen T, Gohlke P, Culman J. Activation of cerebral peroxisome prolif-erator-activated receptors gamma promotes neuroprotection by attenuation of neuronal cyclooxygenase-2 overexpression after focal cerebral ischemia in rats. FASEB J. 2006;20:1162–1175. doi: 10.1096/fj.05-5007com. [DOI] [PubMed] [Google Scholar]
  • 55.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 PPARgamma 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]
  • 56.Sundararajan S, Gamboa JL, Victor NA, Wanderi EW, Lust WD, Landreth GE. Peroxisome proliferator-activated receptor-gamma ligands reduce inflammation and infarction size in transient focal ischemia. Neuroscience. 2005;130:685–696. doi: 10.1016/j.neuroscience.2004.10.021. [DOI] [PubMed] [Google Scholar]
  • 57.Pereira MP, Hurtado O, Cardenas A, onso-Escolano D, Bosca L, Vivancos J, Nombela F, Leza JC, Lorenzo P, Lizasoain I, Moro MA. The nonthiazolidinedione PPARgamma agonist L-796,449 is neuroprotective in experimental stroke. J Neuropathol Exp Neurol. 2005;64:797–805. doi: 10.1097/01.jnen.0000178852.83680.3c. [DOI] [PubMed] [Google Scholar]
  • 58.Shimazu T, Inoue I, Araki N, Asano Y, Sawada M, Furuya D, Nagoya H, Greenberg JH. A peroxisome proliferator-activated receptor-gamma agonist reduces infarct size in transient but not in permanent ischemia. Stroke. 2005;36:353–359. doi: 10.1161/01.STR.0000152271.21943.a2. [DOI] [PubMed] [Google Scholar]
  • 59.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]
  • 60.Savitz SI, Rosenbaum DM. Apoptosis in neurological disease. Neurosurgery. 1998;42:555–572. doi: 10.1097/00006123-199803000-00026. [DOI] [PubMed] [Google Scholar]
  • 61.Wong J, Hoe NW, Zhiwei F, Ng I. Apoptosis and traumatic brain injury. Neurocrit Care. 2005;3:177–182. doi: 10.1385/NCC:3:2:177. [DOI] [PubMed] [Google Scholar]
  • 62.Berger RP, Pierce MC, Wisniewski SR, Adelson PD, Clark RS, Ruppel RA, Kochanek PM. Neuron-specific enolase and S100B in cerebrospinal fluid after severe traumatic brain injury in infants and children. Pediatrics. 2002;109:E31. doi: 10.1542/peds.109.2.e31. [DOI] [PubMed] [Google Scholar]
  • 63.Luo C, Jiang J, Lu Y, Zhu C. Spatial and temporal profile of apoptosis following lateral fluid percussion brain injury. Chin J Traumatol. 2002;5:24–27. [PubMed] [Google Scholar]
  • 64.O'Dell DM, Raghupathi R, Crino PB, Eberwine JH, McIntosh TK. Traumatic brain injury alters the molecular fingerprint of TUNEL-positive cortical neurons In vivo: A single-cell analysis. J Neu-rosci. 2000;20:4821–4828. doi: 10.1523/JNEUROSCI.20-13-04821.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Raghupathi R, Graham DI, McIntosh TK. Apoptosis after traumatic brain injury. J Neurotrauma. 2000;17:927–938. doi: 10.1089/neu.2000.17.927. [DOI] [PubMed] [Google Scholar]
  • 66.Knoblach SM, Nikolaeva M, Huang X, Fan L, Krajewski S, Reed JC, Faden AI. Multiple caspases are activated after traumatic brain injury: evidence for involvement in functional outcome. J Neurotrauma. 2002;19:1155–1170. doi: 10.1089/08977150260337967. [DOI] [PubMed] [Google Scholar]
  • 67.LarnerSF, McKinsey DM, Hayes RL, KK WW. Caspase 7: increased expression and activation after traumatic brain injury in rats. J Neurochem. 2005;94:97–108. doi: 10.1111/j.1471-4159.2005.03172.x. [DOI] [PubMed] [Google Scholar]
  • 68.Larner SF, Hayes RL, McKinsey DM, Pike BR, Wang KK. Increased expression and processing of caspase-12 after traumatic brain injury in rats. J Neurochem. 2004;88:78–90. doi: 10.1046/j.1471-4159.2003.02141.x. [DOI] [PubMed] [Google Scholar]
  • 69.Clark RS, Kochanek PM, Chen M, Watkins SC, Marion DW, Chen J, Hamilton RL, Loeffert JE, Graham SH. Increases in Bcl-2 and cleavage of caspase-1 and caspase-3 in human brain after head injury. FASEBJ. 1999;13:813–821. doi: 10.1096/fasebj.13.8.813. [DOI] [PubMed] [Google Scholar]
  • 70.Dressler J, Hanisch U, Kuhlisch E, Geiger KD. Neuronal and glial apoptosis in human traumatic brain injury. Int J Legal Med. 2007;121:365–375. doi: 10.1007/s00414-006-0126-6. [DOI] [PubMed] [Google Scholar]
  • 71.Zhang X, Graham SH, Kochanek PM, Marion DW, Nathaniel PD, Watkins SC, Clark RS. Caspase-8 expression and proteolysis in human brain after severe head injury. FASEB J. 2003;17:1367–1369. doi: 10.1096/fj.02-1067fje. [DOI] [PubMed] [Google Scholar]
  • 72.Dehmer T, Heneka MT, Sastre M, Dichgans J, Schulz JB. Protection by pioglitazone in the MPTP model of Parkinson's disease correlates with I kappa B alpha induction and block of NF kappa B and iNOS activation. J Neurochem. 2004;88:494–501. doi: 10.1046/j.1471-4159.2003.02210.x. [DOI] [PubMed] [Google Scholar]
  • 73.Breidert T, Callebert J, Heneka MT, Landreth G, Launay JM, Hirsch EC. Protective action of the peroxisome proliferator-activated receptor-gamma agonist pioglitazone in a mouse model of Parkinson's disease. J Neurochem. 2002;82:615–624. doi: 10.1046/j.1471-4159.2002.00990.x. [DOI] [PubMed] [Google Scholar]
  • 74.Heneka MT, Klockgether T, Feinstein DL. Peroxisome proliferator-activated receptor-gamma ligands reduce neuronal inducible nitric oxide synthase expression and cell death in vivo. J Neurosci. 2000;20:6862–6867. doi: 10.1523/JNEUROSCI.20-18-06862.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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]
  • 76.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]
  • 77.Zhao X, Ou Z, Grotta JC, Waxham N, Aronowski J. Peroxisome-proliferator-activated receptor-gamma (PPARgamma) activation protects neurons from NMDA excitotoxicity. Brain Res. 2006;1073-1074:460–469. doi: 10.1016/j.brainres.2005.12.061. [DOI] [PubMed] [Google Scholar]
  • 78.Morosetti R, Servidei T, Mirabella M, Rutella S, Mangiola A, Maira G, Mastrangelo R, Koeffler HP. The PPARgamma ligands PGJ2 and rosiglitazone show a differential ability to inhibit proliferation and to induce apoptosis and differentiation of human glioblastoma cell lines. Int J Oncol. 2004;25:493–502. [PubMed] [Google Scholar]
  • 79.Chattopadhyay N, Singh DP, Heese O, Godbole MM, Sinohara T, Black PM, Brown EM. Expression of peroxisome proliferator-activated receptors (PPARS) in human astrocytic cells: PPARgamma agonists as inducers of apoptosis. J Neurosci Res. 2000;61:67–74. doi: 10.1002/1097-4547(20000701)61:1<67::AID-JNR8>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 80.Zander T, Kraus JA, Grommes C, Schlegel U, Feinstein D, Klockgether T, Landreth G, Koenigsknecht J, Heneka MT. Induction of apoptosis in human and rat glioma by agonists of the nuclear receptor PPARgamma. J Neurochem. 2002;81:1052–1060. doi: 10.1046/j.1471-4159.2002.00899.x. [DOI] [PubMed] [Google Scholar]
  • 81.Servidei T, Morosetti R, Ferlini C, Cusano G, Scambia G, Mastrangelo R, Koeffler HP. The cellular response to PPARgamma ligands is related to the phenotype of neuroblastoma cell lines. Oncol Res. 2004;14:345–354. doi: 10.3727/0965040041292297. [DOI] [PubMed] [Google Scholar]
  • 82.Rodway HA, Hunt AN, Kohler JA, Postle AD, Lillycrop KA. Lysophosphatidic acid attenuates the cytotoxic effects and degree of peroxisome proliferator-activated receptor gamma activation induced by 15-deoxyDelta12,14-prostaglandin J2 in neuroblastoma cells. Biochem J. 2004;382:83–91. doi: 10.1042/BJ20040107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Na HK, Surh YJ. Peroxisome proliferator-activated receptor gamma (PPARgamma) ligands as bifunctional regulators of cell proliferation. Biochem Pharmacol. 2003;66:1381–1391. doi: 10.1016/s0006-2952(03)00488-x. [DOI] [PubMed] [Google Scholar]
  • 84.Clay CE, Namen AM, Atsumi G, Willingham MC, High KP, Kute TE, Trimboli AJ, Fonteh AN, Dawson PA, Chilton FH. Influence of J series prostaglandins on apoptosis and tumorigenesis of breast cancer cells. Carcinogenesis. 1999;20:1905–1911. doi: 10.1093/carcin/20.10.1905. [DOI] [PubMed] [Google Scholar]
  • 85.Louissaint A, Jr, Rao S, Leventhal C, Goldman SA. Coordinated interaction of neurogene-sis and angiogenesis in the adult songbird brain. Neuron. 2002;34:945–960. doi: 10.1016/s0896-6273(02)00722-5. [DOI] [PubMed] [Google Scholar]
  • 86.Barami K. Relationship of neural stem cells with their vascular niche: implications in the malignant progression of gliomas. J Clin Neurosci. 2008;15:1193–1197. doi: 10.1016/j.jocn.2008.01.002. [DOI] [PubMed] [Google Scholar]
  • 87.Curtis MA, Kam M, Nannmark U, Anderson MF, Axell MZ, Wikkelso C, Holtas S, van Roon-Mom WM, Bjork-Eriksson T, Nordborg C, Frisen J, Dragunow M, Faull RL, Eriksson PS. Human neuroblasts migrate to the olfactory bulb via a lateral ventricular extension. Science. 2007;315:1243–1249. doi: 10.1126/science.1136281. [DOI] [PubMed] [Google Scholar]
  • 88.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]
  • 89.varez-Buylla A, Garcia-Verdugo JM, Tramontin AD. A unified hypothesis on the lineage of neural stem cells. Nat Rev Neurosci. 2001;2:287–293. doi: 10.1038/35067582. [DOI] [PubMed] [Google Scholar]
  • 90.Temple S, Qian X. bFGF, neurotrophins, and the control or cortical neurogenesis. Neuron. 1995;15:249–252. doi: 10.1016/0896-6273(95)90030-6. [DOI] [PubMed] [Google Scholar]
  • 91.Kernie SG, Parent JM. Forebrain neurogenesis after focal Ischemic and traumatic brain injury. Neurobiol Dis. 2010;37:267–274. doi: 10.1016/j.nbd.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Anderson VA, Catroppa C, Rosenfeld J, Haritou F, Morse SA. Recovery of memory function following traumatic brain injury in pre-school children. Brain Inj. 2000;14:679–692. doi: 10.1080/026990500413704. [DOI] [PubMed] [Google Scholar]
  • 93.Demeurisse G. Mechanisms of functional restoration after brain injury. Acta Neurol Belg. 2000;100:77–83. [PubMed] [Google Scholar]
  • 94.Ewing-Cobbs L, Barnes MA, Fletcher JM. Early brain injury in children: development and reorganization of cognitive function. Dev Neuropsychol. 2003;24:669–704. doi: 10.1080/87565641.2003.9651915. [DOI] [PubMed] [Google Scholar]
  • 95.Chen XH, Iwata A, Nonaka M, Browne KD, Smith DH. Neurogenesis and glial proliferation persist for at least one year in the subven-tricular zone following brain trauma in rats. J Neurotrauma. 2003;20:623–631. doi: 10.1089/089771503322144545. [DOI] [PubMed] [Google Scholar]
  • 96.Urrea C, Castellanos DA, Sagen J, Tsoulfas P, Bramlett HM, Dietrich WD. Widespread cellular proliferation and focal neurogenesis after traumatic brain injury in the rat. Restor Neurol Neurosci. 2007;25:65–76. [PubMed] [Google Scholar]
  • 97.Chirumamilla S, Sun D, Bullock MR, Colello RJ. Traumatic brain injury induced cell proliferation in the adult mammalian central nervous system. J Neurotrauma. 2002;19:693–703. doi: 10.1089/08977150260139084. [DOI] [PubMed] [Google Scholar]
  • 98.Katayama K, Wada K, Nakajima A, Kamisaki Y, Mayumi T. Nuclear receptors as targets for drug development: the role of nuclear receptors during neural stem cell proliferation and differentiation. J Pharmacol Sci. 2005;97:171–176. doi: 10.1254/jphs.fmj04008x3. [DOI] [PubMed] [Google Scholar]
  • 99.Braissant O, Wahli W. Differential expression of peroxisome proliferator-activated receptor-alpha, -beta, and -gamma during rat embryonic development. Endocrinol. 1998;139:2748–2754. doi: 10.1210/endo.139.6.6049. [DOI] [PubMed] [Google Scholar]
  • 100.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]
  • 101.Gensch C, Clever YP, Werner C, Hanhoun M, Bohm M, Laufs U. The PPAR-gamma agonist pioglitazone increases neoangiogenesis and prevents apoptosis of endothelial progenitor cells. Atherosclerosis. 2007;192:67–74. doi: 10.1016/j.atherosclerosis.2006.06.026. [DOI] [PubMed] [Google Scholar]
  • 102.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]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES