期刊论文详细信息
Biology of Sex Differences
Sex differences in the inflammatory response of primary astrocytes to lipopolysaccharide
María Santos-Galindo1  Estefanía Acaz-Fonseca1  María J Bellini1  Luis M Garcia-Segura1 
[1] Instituto Cajal, CSIC, Avenida Doctor Arce 37, E-28002 Madrid, Spain
关键词: TNFα;    translocator protein 18 kDa;    Toll-like receptor 4;    testosterone;    steroidogenic acute regulatory protein;    IL6;    IL1β;    IFN-inducible protein 10;   
Others  :  793942
DOI  :  10.1186/2042-6410-2-7
 received in 2011-04-04, accepted in 2011-07-11,  发布年份 2011
PDF
【 摘 要 】

Background

Numerous neurological and psychiatric disorders show sex differences in incidence, age of onset, symptomatology or outcome. Astrocytes, one of the glial cell types of the brain, show sex differences in number, differentiation and function. Since astrocytes are involved in the response of neural tissue to injury and inflammation, these cells may participate in the generation of sex differences in the response of the brain to pathological insults. To explore this hypothesis, we have examined whether male and female astrocytes show a different response to an inflammatory challenge and whether perinatal testosterone influences this response.

Methods

Cortical astrocyte cultures were prepared from postnatal day 1 (one day after birth) male or female CD1 mice pups. In addition, cortical astrocyte cultures were also prepared from female pups that were injected at birth with 100 μg of testosterone propionate or vehicle. Cultures were treated for 5 hours with medium containing lipopolysaccharide (LPS) or with control medium. The mRNA levels of IL6, interferon-inducible protein 10 (IP10), TNFα, IL1β, Toll-like receptor 4 (TLR4), steroidogenic acute regulatory protein and translocator protein were assessed by quantitative real-time polymerase chain reaction. Statistical significance was assessed by unpaired t-test or by one-way analysis of variance followed by the Tukey post hoc test.

Results

The mRNA levels of IL6, TNFα and IL1β after LPS treatment were significantly higher in astrocytes derived from male or androgenized females compared to astrocytes derived from control or vehicle-injected females. In contrast, IP10 mRNA levels after LPS treatment were higher in astrocytes derived from control or vehicle-injected females than in those obtained from males or androgenized females. The different response of male and female astrocytes to LPS was due neither to differences in the basal expression of the inflammatory molecules nor to differences in the expression of the LPS receptor TLR4. In contrast, the different inflammatory response was associated with increased mRNA levels of translocator protein, a key steroidogenic regulator, in female astrocytes that were treated with LPS.

Conclusions

Male and female cortical astrocytes respond differentially to an inflammatory challenge and this may be predetermined by perinatal testosterone exposure.

【 授权许可】

   
2011 Santos-Galindo et al; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140705061243609.pdf 354KB PDF download
Figure 6. 20KB Image download
Figure 5. 26KB Image download
Figure 4. 41KB Image download
Figure 3. 33KB Image download
Figure 2. 37KB Image download
Figure 1. 30KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

【 参考文献 】
  • [1]Magistretti PJ: Neuron-glia metabolic coupling and plasticity. J Exp Biol 2006, 209:2304-2311.
  • [2]Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, Newman EA: Glial and neuronal control of brain blood flow. Nature 2010, 468:232-243.
  • [3]Gundersen V, Volterra A: Glutamate exocytosis from astrocytes controls synaptic strength. Nat Neurosci 2007, 10:331-339.
  • [4]Araque A, Navarrete M: Glial cells in neuronal network function. Philos Trans R Soc Lond B Biol Sci 2010, 365:2375-2381.
  • [5]Cerciat M, Unkila M, Garcia-Segura LM, Arevalo MA: Selective estrogen receptor modulators decrease the production of interleukin-6 and interferon-γ-inducible protein-10 by astrocytes exposed to inflammatory challenge in vitro. Glia 2010, 58:93-102.
  • [6]Bellini MJ, Hereñú CB, Goya RG, Garcia-Segura LM: Insulin-like growth factor-I gene delivery to astrocytes reduces their inflammatory response to lipopolysaccharide. J Neuroinflammation 2011, 8:21. BioMed Central Full Text
  • [7]Dong Y, Benveniste EN: Immune function of astrocytes. Glia 2001, 36:180-190.
  • [8]Blanco AM, Valles SL, Pascual M, Guerri C: Involvement of TLR4/type I IL-1 receptor signaling in the induction of inflammatory mediators and cell death induced by ethanol in cultured astrocytes. J Immunol 2005, 175:6893-6899.
  • [9]Krasowska-Zoladek A, Banaszewska M, Kraszpulski M, Konat GW: Kinetics of inflammatory response of astrocytes induced by TLR 3 and TLR4 ligation. J Neurosci Res 2007, 85:205-212.
  • [10]Konat GW, Krasowska-Zoladek A, Kraszpulski M: Statins enhance toll-like receptor 4-mediated cytokine gene expression in astrocytes: implication of Rho proteins in negative feedback regulation. J Neurosci Res 2008, 86:603-609.
  • [11]Liao CK, Wang SM, Chen YL, Wang HS, Wu JC: Lipopolysaccharide-induced inhibition of connexin43 gap junction communication in astrocytes is mediated by downregulation of caveolin-3. Int J Biochem Cell Biol 2010, 42:762-770.
  • [12]Gorina R, Font-Nieves M, Márquez-Kisinousky L, Santalucia T, Planas AM: Astrocyte TLR4 activation induces a proinflammatory environment through the interplay between MyD88-dependent NFκB signaling, MAPK, and Jak1/Stat1 pathways. Glia 2011, 59:242-255.
  • [13]Amateau SK, McCarthy MM: Sexual differentiation of astrocyte morphology in the developing rat preoptic area. J Neuroendocrinol 2002, 14:904-910.
  • [14]Suárez I, Bodega G, Rubio M, Fernandez B: Sexual dimorphism in the distribution of glial fibrillary acidic protein in the supraoptic nucleus of the hamster. J Anat 1991, 178:79-82.
  • [15]Garcia-Segura LM, Dueñas M, Busiguina S, Naftolin F, Chowen JA: Gonadal hormone regulation of neuronal-glial interactions in the developing neuroendocrine hypothalamus. J Steroid Biochem Mol Biol 1995, 53:293-298.
  • [16]Collado P, Beyer C, Hutchison JB, Holman SD: Hypothalamic distribution of astrocytes is gender-related in Mongolian gerbils. Neurosci Lett 1995, 184:86-89.
  • [17]Mong JA, McCarthy MM: Steroid-induced developmental plasticity in hypothalamic astrocytes: Implications for synaptic patterning. J Neurobiol 1999, 40:602-619.
  • [18]Kuo J, Hamid N, Bondar G, Dewing P, Clarkson J, Micevych P: Sex differences in hypothalamic astrocyte response to estradiol stimulation. Biol Sex Differ 2010, 1:7. BioMed Central Full Text
  • [19]Suárez I, Bodega G, Rubio M, Fernández B: Sexual dimorphism in the hamster cerebellum demonstrated by glial fibrillary acidic protein (GFAP) and vimentin immunoreactivity. Glia 1992, 5:10-16.
  • [20]Garcia-Segura LM, Suarez I, Segovia S, Tranque PA, Calés JM, Aguilera P, Olmos G, Guillamón A: The distribution of glial fibrillary acidic protein in the adult rat brain is influenced by the neonatal levels of sex steroids. Brain Res 1988, 456:357-363.
  • [21]Rasia-Filho AA, Xavier LL, dos Santos P, Gehlen G, Achaval M: Glial fibrillary acidic protein immunodetection and immunoreactivity in the anterior and posterior medial amygdala of male and female rats. Brain Res Bull 2002, 58:67-75.
  • [22]Johnson RT, Breedlove SM, Jordan CL: Sex differences and laterality in astrocyte number and complexity in the adult rat medial amygdala. J Comp Neurol 2008, 511:599-609.
  • [23]Conejo NM, González-Pardo H, Cimadevilla JM, Argüelles JA, Díaz F, Vallejo-Seco G, Arias JL: Influence of gonadal steroids on the glial fibrillary acidic protein-immunoreactive astrocyte population in young rat hippocampus. J Neurosci Res 2005, 79:488-494.
  • [24]Arias C, Zepeda A, Hernández-Ortega K, Leal-Galicia P, Lojero C, Camacho-Arroyo I: Sex and estrous cycle-dependent differences in glial fibrillary acidic protein immunoreactivity in the adult rat hippocampus. Horm Behav 2009, 55:257-263.
  • [25]Garcia-Segura LM, Chowen JA, Párducz A, Naftolin F: Gonadal hormones as promoters of structural synaptic plasticity: cellular mechanisms. Prog Neurobiol 1994, 44:279307.
  • [26]Garcia-Segura LM: Hormones and Brain Plasticity. New York: Oxford University Press; 2009.
  • [27]Azad NA, Al Bugami M, Loy-English I: Gender differences in dementia risk factors. Gend Med 2007, 4:120-129.
  • [28]Figueira ML, Ouakinin S: Gender-related endocrinological dysfunction and mental disorders. Curr Opin Psychiatry 2010, 23:369-372.
  • [29]Jobin C, Larochelle C, Parpal H, Coyle PK, Duquette P: Gender issues in multiple sclerosis: an update. Womens Health (Lond Engl) 2010, 6:797-820.
  • [30]Melcangi RC, Garcia-Segura LM: Sex-specific therapeutic strategies based on neuroactive steroids: in search for innovative tools for neuroprotection. Horm Behav 2010, 57:2-11.
  • [31]Voskuhl R: Sex differences in autoimmune diseases. Biol Sex Differ 2011, 2:1. BioMed Central Full Text
  • [32]Liu M, Hurn PD, Roselli CE, Alkayed NJ: Role of P450 aromatase in sex-specific astrocytic cell death. J Cereb Blood Flow Metab 2007, 27:135-141.
  • [33]Garcia-Segura LM, Veiga S, Sierra A, Melcangi RC, Azcoitia I: Aromatase: a neuroprotective enzyme. Prog Neurobiol 2003, 71:31-41.
  • [34]Saldanha CJ, Duncan KA, Walters BJ: Neuroprotective actions of brain aromatase. Front Neuroendocrinol 2009, 30:106-118.
  • [35]Garcia-Segura LM, Melcangi RC: Steroids and glial cell function. Glia 2006, 54:485-498.
  • [36]Sinchak K, Mills RH, Tao L, LaPolt P, Lu JK, Micevych P: Estrogen induces de novo progesterone synthesis in astrocytes. Dev Neurosci 2003, 25:343-348.
  • [37]Veiga S, Garcia-Segura LM, Azcoitia I: Neuroprotection by the steroids pregnenolone and dehydroepiandrosterone is mediated by the enzyme aromatase. J Neurobiol 2003, 56:398-406.
  • [38]Ciriza I, Azcoitia I, Garcia-Segura LM: Reduced progesterone metabolites protect rat hippocampal neurones from kainic acid excitotoxicity in vivo. J Neuroendocrinol 2004, 16:58-63.
  • [39]De Nicola AF, Labombarda F, Deniselle MC, Gonzalez SL, Garay L, Meyer M, Gargiulo G, Guennoun R, Schumacher M: Progesterone neuroprotection in traumatic CNS injury and motoneuron degeneration. Front Neuroendocrinol 2009, 30:173-187.
  • [40]Stein DG, Wright DW: Progesterone in the clinical treatment of acute traumatic brain injury. Expert Opin Investig Drugs 2010, 19:847-857.
  • [41]Moralí G, Montes P, Hernández-Morales L, Monfil T, Espinosa-García C, Cervantes M: Neuroprotective effects of progesterone and allopregnanolone on long-term cognitive outcome after global cerebral ischemia. Restor Neurol Neurosci 2011, 29:1-15.
  • [42]Dang J, Mitkari B, Kipp M, Beyer C: Gonadal steroids prevent cell damage and stimulate behavioral recovery after transient middle cerebral artery occlusion in male and female rats. Brain Behav Immun 2011, 25:715-726.
  • [43]Itzhak Y, Roig-Cantisano A, Norenberg MD: Ontogeny of peripheral-type benzodiazepine receptors in cultured astrocytes and brain from rat. Brain Res Dev Brain Res 1995, 84:62-66.
  • [44]Sierra A: Neurosteroids: the StAR protein in the brain. J Neuroendocrinol 2004, 16:787-793.
  • [45]Lavaque E, Sierra A, Azcoitia I, Garcia-Segura LM: Steroidogenic acute regulatory protein in the brain. Neuroscience 2006, 138:741-747.
  • [46]Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapère JJ, Lindemann P, Norenberg MD, Nutt D, Weizman A, Zhang MR, Gavish M: Translocator protein (18 kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci 2006, 27:402-409.
  • [47]Arnold AP, Gorski RA: Gonadal steroid induction of structural sex differences in the central nervous system. Annu Rev Neurosci 1984, 7:413-442.
  • [48]Pistritto G, Franzese O, Pozzoli G, Mancuso C, Tringali G, Preziosi P, Navarra P: Bacterial lipopolysaccharide increases prostaglandin production by rat astrocytes via inducible cyclo-oxygenase: Evidence for the involvement of nuclear factor κB. Biochem Biophys Res Commun 1999, 263:570-574.
  • [49]Caruso D, D'Intino G, Giatti S, Maschi O, Pesaresi M, Calabrese D, Garcia-Segura LM, Calza L, Melcangi RC: Sex-dimorphic changes in neuroactive steroid levels after chronic experimental autoimmune encephalomyelitis. J Neurochem 2010, 114:921-932.
  • [50]Pesaresi M, Maschi O, Giatti S, Garcia-Segura LM, Caruso D, Melcangi RC: Sex differences in neuroactive steroid levels in the nervous system of diabetic and non-diabetic rats. Horm Behav 2010, 57:46-55.
  • [51]Rone MB, Fan J, Papadopoulos V: Cholesterol transport in steroid biosynthesis: role of protein-protein interactions and implications in disease states. Biochim Biophys Acta 2009, 1791:646-658.
  • [52]Garcia-Estrada J, Del Rio JA, Luquin S, Soriano E, Garcia-Segura LM: Gonadal hormones down-regulate reactive gliosis and astrocyte proliferation after a penetrating brain injury. Brain Res 1993, 628:271-278.
  • [53]García-Estrada J, Luquín S, Fernández AM, Garcia-Segura LM: Dehydroepiandrosterone, pregnenolone and sex steroids down-regulate reactive astroglia in the male rat brain after a penetrating brain injury. Int J Dev Neurosci 1999, 17:145-151.
  • [54]Grossman KJ, Goss CW, Stein DG: Effects of progesterone on the inflammatory response to brain injury in the rat. Brain Res 2004, 1008:29-39.
  • [55]Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D: Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci 2007, 25:3039-3046.
  • [56]Gehlert DR, Stephenson DT, Schober DA, Rash K, Clemens JA: Increased expression of peripheral benzodiazepine receptors in the facial nucleus following motor neuron axotomy. Neurochem Int 1997, 31:705-713.
  • [57]Rojas S, Martín A, Arranz MJ, Pareto D, Purroy J, Verdaguer E, Llop J, Gómez V, Gispert JD, Millán O, Chamorro A, Planas AM: Imaging brain inflammation with [11C]PK11195 by PET and induction of the peripheral-type benzodiazepine receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab 2007, 27:1975-1986.
  • [58]Chen MK, Guilarte TR: Translocator protein 18 kDa (TSPO): molecular sensor of brain injury and repair. Pharmacol Ther 2008, 118:1-17.
  • [59]Veiga S, Azcoitia I, Garcia-Segura LM: Ro5-4864, a peripheral benzodiazepine receptor ligand, reduces reactive gliosis and protects hippocampal hilar neurons from kainic acid excitotoxicity. J Neurosci Res 2005, 80:129-137.
  • [60]Veiga S, Carrero P, Pernia O, Azcoitia I, Garcia-Segura LM: Translocator protein 18 kDa is involved in the regulation of reactive gliosis. Glia 2007, 55:1426-1436.
  • [61]Veenman L, Papadopoulos V, Gavish M: Channel-like functions of the 18-kDa translocator protein (TSPO): regulation of apoptosis and steroidogenesis as part of the host-defense response. Curr Pharm Des 2007, 13:2385-2405.
  • [62]Veenman L, Shandalov Y, Gavish M: VDAC activation by the 18 kDa translocator protein (TSPO), implications for apoptosis. J Bioenerg Biomembr 2008, 40:199-205.
  • [63]Sierra A, Lavaque E, Perez-Martin M, Azcoitia I, Hales DB, Garcia-Segura LM: Steroidogenic acute regulatory protein in the rat brain: cellular distribution, developmental regulation and overexpression after injury. Eur J Neurosci 2003, 18:1458-1467.
  • [64]Lavaque E, Mayen A, Azcoitia I, Tena-Sempere M, Garcia-Segura LM: Sex differences, developmental changes, response to injury and cAMP regulation of the mRNA levels of steroidogenic acute regulatory protein, cytochrome p450scc, and aromatase in the olivocerebellar system. J Neurobiol 2006, 66:308-318.
  • [65]Arnold AP: The organizational-activational hypothesis as the foundation for a unified theory of sexual differentiation of all mammalian tissues. Horm Behav 2009, 55:570-578.
  • [66]Mong JA, Glaser E, McCarthy MM: Gonadal steroids promote glial differentiation and alter neuronal morphology in the developing hypothalamus in a regionally specific manner. J Neurosci 1999, 19:1464-1472.
  • [67]Liu M, Oyarzabal EA, Yang R, Murphy SJ, Hurn PD: A novel method for assessing sex-specific and genotype-specific response to injury in astrocyte culture. J Neurosci Methods 2008, 171:214-217.
  • [68]Farber JM: Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol 1997, 61:246-257.
  • [69]Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD: IFN-γ-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol 2002, 168:3195-3204.
  • [70]Rothwell N: Interleukin-1 and neuronal injury: mechanisms, modification, and therapeutic potential. Brain Behav Immun 2003, 17:152-157.
  • [71]Mason JL, Suzuki K, Chaplin DD, Matsushima GK: Interleukin-1β promotes repair of the CNS. J Neurosci 2001, 21:7046-7052.
  • [72]Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MBA, Mucke L: Neurologic disease in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci USA 1993, 90:10061-10065.
  • [73]Swartz KR, Liu F, Sewell D, Schochet T, Campbell I, Sandor M, Fabry Z: Interleukin-6 promotes post-traumatic healing in the central nervous system. Brain Res 2001, 896:86-95.
  • [74]Penkowa M, Giralt M, Lago N, Camats J, Carrasco J, Hernandez J, Molinero A, Campbell IL, Hidalgo J: Astrocyte-targeted expression of IL-6 protects the CNS against a focal brain injury. Exp Neurol 2003, 181:130-148.
  • [75]Quintana A, Molinero A, Borup R, Nielsen FC, Campbell IL, Penkowa M, Hidalgo J: Effect of astrocyte-targeted production of IL-6 on traumatic brain injury and its impact on the cortical transcriptome. Dev Neurobiol 2008, 68:195-208.
  • [76]Syed MM, Phulwani NK, Kielian T: Tumor necrosis factor-α (TNF-α) regulates Toll-like receptor 2 (TLR2) expression in microglia. J Neurochem 2007, 103:1461-1471.
  • [77]Worrall NK, Chang K, LeJeune WS, Misko TP, Sullivan PM, Ferguson TB, Williamson JR: TNF-α causes reversible in vivo systemic vascular barrier dysfunction via NO-dependent and -independent mechanisms. Am J Physiol 1997, 273:H2565-H2574.
  • [78]Kim S, Steelman AJ, Koito H, Li J: Astrocytes promote TNF-mediated toxicity to oligodendrocyte precursors. J Neurochem 2011, 116:53-66.
  • [79]Su Z, Yuan Y, Chen J, Zhu Y, Qiu Y, Zhu F, Huang A, He C: Reactive astrocytes inhibit the survival and differentiation of oligodendrocyte precursor cells by secreted TNF-α. J Neurotrauma 2011, 28:1089-1100.
  文献评价指标  
  下载次数:26次 浏览次数:7次