Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 419
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Ann Neurol ; 90(5): 789-807, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34476836

RESUMEN

OBJECTIVE: Parkinson's disease (PD) manifests in motor dysfunction, non-motor symptoms, and eventual dementia (PDD). Neuropathological hallmarks include nigrostriatal neurodegeneration, Lewy body (LB) pathology, and neuroinflammation. Alpha-synuclein (α-syn), a primary component of LBs, is implicated in PD pathogenesis, accumulating, and aggregating in both familial and sporadic PD. However, as α-syn pathology is often comorbid with amyloid-beta (Aß) plaques and phosphorylated tau (pTau) tangles in PDD, it is still unclear whether α-syn is the primary cause of neurodegeneration in sporadic PDD. We aimed to determine how the absence of α-syn would affect PDD manifestation. METHODS: IFN-ß knockout (Ifnb-/- ) mice spontaneously develop progressive behavior abnormalities and neuropathology resembling PDD, notably with α-syn+ LBs. We generated Ifnb/Snca double knockout (DKO) mice and evaluated their behavior and neuropathology compared with wild-type (Wt), Ifnb-/- , and Snca-/- mice using immunohistochemistry, electron microscopy, immunoblots, qPCR, and modification of neuronal signaling. RESULTS: Ifnb/Snca DKO mice developed all clinical PDD-like behavioral manifestations induced by IFN-ß loss. Independently of α-syn expression, lack of IFN-ß alone induced Aß plaques, pTau tangles, and LB-like Aß+ /pTau+ inclusion bodies and neuroinflammation. IFN-ß loss caused significant elevated glial and neuronal TNF-α and neuronal TNFR1, associated with neurodegeneration. Restoring neuronal IFN-ß signaling or blocking TNFR1 rescued caspase 3/t-BID-mediated neuronal-death through upregulation of c-FLIPS and lowered intraneuronal Aß and pTau accumulation. INTERPRETATION: These findings increase our understanding of PD pathology and suggest that targeting α-syn alone is not sufficient to mitigate disease. Targeting specific aspects of neuroinflammation, such as aberrant neuronal TNF-α/TNFR1 or IFN-ß/IFNAR signaling, may attenuate disease. ANN NEUROL 2021;90:789-807.


Asunto(s)
Enfermedades Neuroinflamatorias/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Progresión de la Enfermedad , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/metabolismo , Ratones Noqueados , Neuroglía/patología , Enfermedad de Parkinson/genética , Placa Amiloide/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia
3.
Alcohol Clin Exp Res ; 44(1): 66-77, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31657476

RESUMEN

BACKGROUND: Chronic alcohol consumption enhances cancer-associated cachexia, which is one of the major causes of decreased survival. The precise molecular mechanism of how alcohol consumption enhances cancer-associated cachexia, especially skeletal muscle loss, remains to be elucidated. METHODS: We used a mouse model of chronic alcohol consumption, in which 20% (w/v) alcohol was provided as sole drinking fluid, and Lewis lung carcinoma to study the underlying mechanisms. RESULTS: We found that alcohol consumption up-regulated the expression of MAFbx, MuRF-1, and LC3 in skeletal muscle, suggesting that alcohol enhanced ubiquitin-mediated proteolysis and LC3-mediated autophagy. Alcohol consumption enhanced phosphorylation of Smad2/3, p38, and ERK and decreased the phosphorylation of FOXO1. These are the signaling molecules governing protein degradation pathways. Moreover, alcohol consumption slightly up-regulated the expression of insulin receptor substrate-1, did not affect phosphatidylinositol-3 kinase, but decreased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), and down-regulated the expression of Raptor and p70 ribosomal kinase S6 kinase, suggesting that alcohol impaired protein synthesis signaling pathway in skeletal muscle of tumor-bearing mice. Alcohol consumption enhanced the expression of myostatin in skeletal muscle, plasma, and tumor, but did not affect the expression of myostatin in non-tumor-bearing mice. In TNFα knockout mice, the effects of alcohol-enhanced expression of myostatin and protein degradation-related signaling molecules, and decreased protein synthesis signaling in skeletal muscle were abolished. Consequently, alcohol consumption neither affected cancer-associated cachexia nor decreased the survival of TNFα KO mice bearing cachectic cancer. CONCLUSIONS: Chronic alcohol consumption enhances cancer-associated skeletal muscle loss through suppressing Akt/mTOR-mediated protein synthesis pathway and enhancing protein degradation pathways. This process is initiated by TNFα and mediated by myostatin.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Etanol/toxicidad , Atrofia Muscular/inducido químicamente , Atrofia Muscular/metabolismo , Miostatina/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia , Animales , Caquexia/inducido químicamente , Caquexia/metabolismo , Etanol/administración & dosificación , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miostatina/antagonistas & inhibidores , Distribución Aleatoria , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
4.
Bull Exp Biol Med ; 169(6): 836-839, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33098510

RESUMEN

Leprosy was modeled in an experiment on BALB/c, BALB/cNude, CBA, and C57BL/6ТNF-/- mice using three Mycobacterium leprae strains obtained from patients with a diagnosis of A30 according to ICD-10 from different regions of the Russian Federation. Proliferation of M. leprae of the used strains showed a temporal-quantitative dependence on the used mouse line. CBA and BALB/cNude mice were optimal for strain R and BALB/c and BALB/cNude lines were optimal for strain I. BALB/cNude mice infected with strain I had low lifespan. M. leprae strain M showed low proliferation activity in BALB/cNude and C57BL/6ТNF-/- mice.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Lepra/inmunología , Longevidad/inmunología , Mycobacterium leprae/patogenicidad , Factor de Necrosis Tumoral alfa/inmunología , Animales , ADN Bacteriano/genética , Modelos Animales de Enfermedad , Especificidad del Huésped , Humanos , Lepra/genética , Lepra/microbiología , Lepra/patología , Longevidad/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Desnudos , Mycobacterium leprae/genética , Mycobacterium leprae/crecimiento & desarrollo , Mycobacterium leprae/inmunología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
5.
Brain Behav Immun ; 82: 279-297, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31505254

RESUMEN

BACKGROUND: Although tumor necrosis factor (TNF) inhibitors are used to treat chronic inflammatory diseases, there is little information about how long-term inhibition of TNF affects the homeostatic functions that TNF maintains in the intact CNS. MATERIALS AND METHODS: To assess whether developmental TNF deficiency causes alterations in the naïve CNS, we estimated the number of proliferating cells, microglia, and neurons in the developing neocortex of E13.5, P7 and adult TNF knock out (TNF-/-) mice and wildtype (WT) littermates. We also measured changes in gene and protein expression and monoamine levels in adult WT and TNF-/- mice. To evaluate long-term effects of TNF inhibitors, we treated healthy adult C57BL/6 mice with either saline, the selective soluble TNF inhibitor XPro1595, or the nonselective TNF inhibitor etanercept. We estimated changes in cell number and protein expression after two months of treatment. We assessed the effects of TNF deficiency on cognition by testing adult WT and TNF-/- mice and mice treated with saline, XPro1595, or etanercept with specific behavioral tasks. RESULTS: TNF deficiency decreased the number of proliferating cells and microglia and increased the number of neurons. At the same time, TNF deficiency decreased the expression of WNT signaling-related proteins, specifically Collagen Triple Helix Repeat Containing 1 (CTHRC1) and Frizzled receptor 6 (FZD6). In contrast to XPro1595, long-term inhibition of TNF with etanercept in adult C57BL/6 mice decreased the number of BrdU+ cells in the granule cell layer of the dentate gyrus. Etanercept, but not XPro1595, also impaired spatial learning and memory in the Barnes maze memory test. CONCLUSION: TNF deficiency impacts the organization of neurogenic zones and alters the cell composition in brain. Long-term inhibition of TNF with the nonselective TNF inhibitor etanercept, but not the soluble TNF inhibitor XPro1595, decreases neurogenesis in the adult mouse hippocampus and impairs learning and memory after two months of treatment.


Asunto(s)
Corteza Cerebral/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia , Animales , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Cognición/efectos de los fármacos , Etanercept/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microglía/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Vía de Señalización Wnt
6.
Arterioscler Thromb Vasc Biol ; 38(2): 457-463, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29217508

RESUMEN

OBJECTIVE: Abdominal aortic aneurysms are inflammatory in nature and are associated with some risk factors that also lead to atherosclerotic occlusive disease, most notably smoking. The purpose of our study was to identify differential cytokine expression in patients with abdominal aortic aneurysm and those with atherosclerotic occlusive disease. Based on this analysis, we further explored and compared the mechanism of action of IL (interleukin)-1ß versus TNF-α (tumor necrosis factor-α) in abdominal aortic aneurysm formation. APPROACH AND RESULTS: IL-1ß was differentially expressed in human plasma with lower levels detected in patients with abdominal aortic aneurysm compared with matched atherosclerotic controls. We further explored its mechanism of action using a murine model and cell culture. Genetic deletion of IL-1ß and IL-1R did not inhibit aneurysm formation or decrease MMP (matrix metalloproteinase) expression. The effects of IL-1ß deletion on M1 macrophage polarization were compared with another proinflammatory cytokine, TNF-α. Bone marrow-derived macrophages from IL-1ß-/- and TNF-α-/- mice were polarized to an M1 phenotype. TNF-α deletion, but not IL-1ß deletion, inhibited M1 macrophage polarization. Infusion of M1 polarized TNF-α-/- macrophages inhibited aortic diameter growth; no inhibitory effect was seen in mice infused with M1 polarized IL-1ß-/- macrophages. CONCLUSIONS: Although IL-1ß is a proinflammatory cytokine, its effects on aneurysm formation and macrophage polarization differ from TNF-α. The differential effects of IL-1ß and TNF-α inhibition are related to M1/M2 macrophage polarization and this may account for the differences in clinical efficacy of IL-1ß and TNF-α antibody therapies in management of inflammatory diseases.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Interleucina-1beta/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Anciano , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/patología , Estudios de Casos y Controles , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-1beta/sangre , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Macrófagos/patología , Macrófagos/trasplante , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fenotipo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
7.
Immunol Invest ; 48(7): 691-703, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30663448

RESUMEN

Introduction: Macrophage phagocytosis of pathogens and tumour cells is an important early event in protection against infectious disease and cancer. As tumour necrosis factor α (TNF) is an important cytokine in macrophage activation, we investigated the involvement of TNF in macrophage phagocytosis of tumour cells. Methods: We used Devil Facial Tumour Disease (DFTD) cancer cells as the target tumour cells. The Tasmanian devil (Sarcophilus harrisii) population is threatened by the transmissible DFTD. Using DFTD cells provided the opportunity to determine if these cells can be phagocytosed and investigate requirement for TNF. As effector cells, bone marrow derived macrophages (BMDMs), generated from C57BL/6 wild type (B6.WT) and C57BL/6 TNF-/- (B6.TNF-/-) mice were used. Phagocytosis of DFTD cells was investigated by confocal microscopy and flow cytometry. Results: DFTD cells were consistently phagocytosed by B6.WT and B6.TNF-/- BMDMs with similar efficiency in vitro. Consequently the DFTD cells are not resistant to phagocytosis. Following activation by exposure to IFNγ and LPS or LPS alone, B6.TNF-/- BMDMs had higher phagocytic efficiency and lower nitric oxide (NO) production compared to wild-type controls. In addition, NO seems to be unlikely to be the involved in phagocytosis efficiency in IFNγ and LPS activated B6.TNF-/- macrophages and consequences thereof. Conclusion: Our results indicate that TNF is not required for IFNγ and LPS or LPS alone activation of macrophage phagocytosis. TNF may negatively regulate macrophage phagocytosis of tumour cells.


Asunto(s)
Neoplasias Faciales/inmunología , Neoplasias Faciales/veterinaria , Macrófagos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Línea Celular Tumoral , Células Cultivadas , Neoplasias Faciales/patología , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Marsupiales , Ratones Endogámicos C57BL , Ratones Mutantes , Óxido Nítrico/metabolismo , Fagocitosis , Factor de Necrosis Tumoral alfa/deficiencia
8.
J Neuroinflammation ; 15(1): 168, 2018 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-29807527

RESUMEN

BACKGROUND: Activated microglia play an essential role in inflammatory responses elicited in the central nervous system (CNS). Microglia-derived extracellular vesicles (EVs) are suggested to be involved in propagation of inflammatory signals and in the modulation of cell-to-cell communication. However, there is a lack of knowledge on the regulation of EVs and how this in turn facilitates the communication between cells in the brain. Here, we characterized microglial EVs under inflammatory conditions and investigated the effects of inflammation on the EV size, quantity, and protein content. METHODS: We have utilized western blot, nanoparticle tracking analysis (NTA), and mass spectrometry to characterize EVs and examine the alterations of secreted EVs from a microglial cell line (BV2) following lipopolysaccharide (LPS) and tumor necrosis factor (TNF) inhibitor (etanercept) treatments, or either alone. The inflammatory responses were measured with multiplex cytokine ELISA and western blot. We also subjected TNF knockout mice to experimental stroke (permanent middle cerebral artery occlusion) and validated the effect of TNF inhibition on EV release. RESULTS: Our analysis of EVs originating from activated BV2 microglia revealed a significant increase in the intravesicular levels of TNF and interleukin (IL)-6. We also observed that the number of EVs released was reduced both in vitro and in vivo when inflammation was inhibited via the TNF pathway. Finally, via mass spectrometry, we identified 49 unique proteins in EVs released from LPS-activated microglia compared to control EVs (58 proteins in EVs released from LPS-activated microglia and 37 from control EVs). According to Gene Ontology (GO) analysis, we found a large increase of proteins related to translation and transcription in EVs from LPS. Importantly, we showed a distinct profile of proteins found in EVs released from LPS treated cells compared to control. CONCLUSIONS: We demonstrate altered EV production in BV2 microglial cells and altered cytokine levels and protein composition carried by EVs in response to LPS challenge. Our findings provide new insights into the potential roles of EVs that could be related to the pathogenesis in neuroinflammatory diseases.


Asunto(s)
Citocinas/metabolismo , Vesículas Extracelulares/patología , Infarto de la Arteria Cerebral Media/complicaciones , Inflamación/etiología , Microglía/patología , Animales , Línea Celular Transformada , Modelos Animales de Enfermedad , Etanercept/farmacología , Vesículas Extracelulares/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Inmunosupresores/farmacología , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
9.
Horm Metab Res ; 50(1): 80-87, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28922680

RESUMEN

Previously, we have shown that the adipocyte-specific nuclear form of sterol regulatory element-binding protein-1c (nSREBP-1c) transgenic mice spontaneously developed hepatic lesions that are similar to those of human nonalcoholic steatohepatitis (NASH) with a concomitant elevation of plasma TNF-α. In this study, we analyzed the role of TNF-α in the progression of nonalcoholic fatty liver disease (NAFLD). We established a Tnf knockout nSREBP-1c transgenic mouse line. Glucose tolerance and liver histology were examined at the age of 20 weeks. The gene expression and protein levels were assessed by quantitative RT-PCR and Western blot, respectively. The Tnf knockout improved glucose tolerance and significantly reduced the prevalence of hepatic steatosis (20% vs. 100%, p<0.0001) and fibrosis (15% vs. 65%, p=0.0057). The expressions of Acaca, Scd1, Mcp1, Tgfb1, Col1a1, and Timp1 were increased in the liver from the original nSREBP-1c transgenic mice. However, gene upregulation was reduced in the livers from the Tnf(-/-) nSREBP-1c transgenic mice. Furthermore, the hepatic levels of TIMP1 protein were increased in the original nSREBP-1c transgenic mice but not in Tnf(-/-) nSREBP-1c transgenic mice. To assess the direct effect of TNF-α on the expression of the genes, we cultured primary hepatocytes in the presence of TNF-α and found that TNF-α increased the expression of Mcp1, Tgfb1, and Timp1 in hepatocytes. These observations indicate that TNF-α plays a pivotal role in the development of NAFLD and progression to NASH through upregulating key molecules associated with lipid metabolism, inflammatory cytokines, and fibrosis in the liver.


Asunto(s)
Progresión de la Enfermedad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hepatocitos/metabolismo , Inflamación/genética , Metabolismo de los Lípidos/genética , Cirrosis Hepática/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia
10.
Nature ; 482(7384): 179-85, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22297845

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome and the leading cause of chronic liver disease in the Western world. Twenty per cent of NAFLD individuals develop chronic hepatic inflammation (non-alcoholic steatohepatitis, NASH) associated with cirrhosis, portal hypertension and hepatocellular carcinoma, yet the causes of progression from NAFLD to NASH remain obscure. Here, we show that the NLRP6 and NLRP3 inflammasomes and the effector protein IL-18 negatively regulate NAFLD/NASH progression, as well as multiple aspects of metabolic syndrome via modulation of the gut microbiota. Different mouse models reveal that inflammasome-deficiency-associated changes in the configuration of the gut microbiota are associated with exacerbated hepatic steatosis and inflammation through influx of TLR4 and TLR9 agonists into the portal circulation, leading to enhanced hepatic tumour-necrosis factor (TNF)-α expression that drives NASH progression. Furthermore, co-housing of inflammasome-deficient mice with wild-type mice results in exacerbation of hepatic steatosis and obesity. Thus, altered interactions between the gut microbiota and the host, produced by defective NLRP3 and NLRP6 inflammasome sensing, may govern the rate of progression of multiple metabolic syndrome-associated abnormalities, highlighting the central role of the microbiota in the pathogenesis of heretofore seemingly unrelated systemic auto-inflammatory and metabolic disorders.


Asunto(s)
Progresión de la Enfermedad , Hígado Graso/metabolismo , Hígado Graso/patología , Inflamasomas/metabolismo , Obesidad/metabolismo , Obesidad/patología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/metabolismo , Colina , Colon/microbiología , Proteínas del Citoesqueleto/deficiencia , Modelos Animales de Enfermedad , Hígado Graso/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-18/deficiencia , Masculino , Metagenoma , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedad del Hígado Graso no Alcohólico , ARN Ribosómico 16S/genética , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/metabolismo
11.
Kidney Blood Press Res ; 42(3): 416-427, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28683439

RESUMEN

BACKGROUND/AIMS: Obese patients and experimental animals exhibit high levels of inflammatory cytokines, such as tumor necrosis factor (TNF)-α. However, the role of TNF-α in the pathophysiologic process in obesity induced kidney damage is still unknown. METHODS: We used TNF-α deficient mice and wild-type (WT) C57/BJ6 mice controls to study the effect of TNF-α on inflammation and oxidative stress in kidney by the model of high-fat diet (HFD) and primary isolated mouse renal proximal tubule cells treated with a mixture of free fatty acids (FFA). RESULTS: Compared with the chow diet group, HFD-fed WT mice had higher urinary albumin and increased levels of renal fibrosis, glomerulosclerosis, inflammation, oxidative stress and apoptosis in the kidney. These changes were co-related with increased expression of TNF-α in the kidney and were attenuated by TNF-α deficiency. In vitro, accumulation of intracellular lipids induced TNF-α expression and oxidative stress in FFA treated primary proximal tubule cells. However, TNF-α inhibition with siRNA or TNF-α deficiency decreased the lipid induced oxidative stress in these cells. CONCLUSION: These findings suggest that TNF-α plays an important role in the HFD induced kidney damage, and targeting TNF-α and/or its receptors could be a promising therapeutic regimen for progressive nephropathy.


Asunto(s)
Nefritis/prevención & control , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/deficiencia , Animales , Dieta Alta en Grasa/efectos adversos , Riñón/lesiones , Lípidos/efectos adversos , Ratones , Obesidad/complicaciones , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
12.
Nature ; 481(7380): 199-203, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22158124

RESUMEN

The largest mucosal surface in the body is in the gastrointestinal tract, a location that is heavily colonized by microbes that are normally harmless. A key mechanism required for maintaining a homeostatic balance between this microbial burden and the lymphocytes that densely populate the gastrointestinal tract is the production and transepithelial transport of poly-reactive IgA (ref. 1). Within the mucosal tissues, B cells respond to cytokines, sometimes in the absence of T-cell help, undergo class switch recombination of their immunoglobulin receptor to IgA, and differentiate to become plasma cells. However, IgA-secreting plasma cells probably have additional attributes that are needed for coping with the tremendous bacterial load in the gastrointestinal tract. Here we report that mouse IgA(+) plasma cells also produce the antimicrobial mediators tumour-necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS), and express many molecules that are commonly associated with monocyte/granulocytic cell types. The development of iNOS-producing IgA(+) plasma cells can be recapitulated in vitro in the presence of gut stroma, and the acquisition of this multifunctional phenotype in vivo and in vitro relies on microbial co-stimulation. Deletion of TNF-α and iNOS in B-lineage cells resulted in a reduction in IgA production, altered diversification of the gut microbiota and poor clearance of a gut-tropic pathogen. These findings reveal a novel adaptation to maintaining homeostasis in the gut, and extend the repertoire of protective responses exhibited by some B-lineage cells.


Asunto(s)
Inmunoglobulina A/inmunología , Intestino Delgado/citología , Intestino Delgado/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Células Cultivadas , Quimera/inmunología , Citrobacter rodentium/inmunología , Técnicas de Cocultivo , Femenino , Vida Libre de Gérmenes , Granulocitos/citología , Granulocitos/metabolismo , Inmunidad Innata/inmunología , Inmunoglobulina A/biosíntesis , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Células Plasmáticas/metabolismo , Bazo/citología , Células del Estroma/citología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Nature ; 471(7340): 591-6, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21455173

RESUMEN

Members of the tumour necrosis factor (TNF) receptor superfamily have important functions in immunity and inflammation. Recently linear ubiquitin chains assembled by a complex containing HOIL-1 and HOIP (also known as RBCK1 and RNF31, respectively) were implicated in TNF signalling, yet their relevance in vivo remained uncertain. Here we identify SHARPIN as a third component of the linear ubiquitin chain assembly complex, recruited to the CD40 and TNF receptor signalling complexes together with its other constituents, HOIL-1 and HOIP. Mass spectrometry of TNF signalling complexes revealed RIP1 (also known as RIPK1) and NEMO (also known as IKKγ or IKBKG) to be linearly ubiquitinated. Mutation of the Sharpin gene (Sharpin(cpdm/cpdm)) causes chronic proliferative dermatitis (cpdm) characterized by inflammatory skin lesions and defective lymphoid organogenesis. Gene induction by TNF, CD40 ligand and interleukin-1ß was attenuated in cpdm-derived cells which were rendered sensitive to TNF-induced death. Importantly, Tnf gene deficiency prevented skin lesions in cpdm mice. We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced cell death and, thereby, prevents inflammation. Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating immune signalling.


Asunto(s)
Inmunidad/inmunología , Inflamación/metabolismo , Transducción de Señal , Ubiquitinación , Animales , Ligando de CD40/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular , Humanos , Quinasa I-kappa B/metabolismo , Inflamación/patología , Inflamación/prevención & control , Interleucina-1beta/metabolismo , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Piel/citología , Piel/inmunología , Piel/metabolismo , Piel/patología , Factores de Transcripción , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Ubiquitina/química , Ubiquitina/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/química , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
14.
Carcinogenesis ; 37(1): 72-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26586792

RESUMEN

Protein kinase C epsilon (PKCε), a Ca(2+)-independent phospholipid-dependent serine/threonine kinase, is among the six PKC isoforms (α, δ, ε, η, µ, ζ) expressed in both mouse and human skin. Epidermal PKCε level dictates the susceptibility of PKCε transgenic (TG) mice to the development of cutaneous squamous cell carcinomas (SCC) elicited either by repeated exposure to ultraviolet radiation (UVR) or by using the DMBA initiation-TPA (12-O-tetradecanoylphorbol-13-acetate) tumor promotion protocol (Wheeler,D.L. et al. (2004) Protein kinase C epsilon is an endogenous photosensitizer that enhances ultraviolet radiation-induced cutaneous damage and development of squamous cell carcinomas. Cancer Res., 64, 7756-7765). Histologically, SCC in TG mice, like human SCC, is poorly differentiated and metastatic. Our earlier studies to elucidate mechanisms of PKCε-mediated development of SCC, using either DMBA-TPA or UVR, indicated elevated release of cytokine TNFα. To determine whether TNFα is essential for the development of SCC in TG mice, we generated PKCε transgenic mice/TNFα-knockout (TG/TNFαKO) by crossbreeding TNFαKO with TG mice. We now present that deletion of TNFα in TG mice inhibited the development of SCC either by repeated UVR exposures or by the DMBA-TPA protocol. TG mice deficient in TNFα elicited both increase in SCC latency and decrease in SCC incidence. Inhibition of UVR-induced SCC development in TG/TNFαKO was accompanied by inhibition of (i) the expression levels of TNFα receptors TNFRI and TNFRII and cell proliferation marker ornithine decarboxylase and metastatic markers MMP7 and MMP9, (ii) the activation of transcription factors Stat3 and NF-kB and (iii) proliferation of hair follicle stem cells and epidermal hyperplasia. The results presented here provide the first genetic evidence that TNFα is linked to PKCε-mediated sensitivity to DMBA-TPA or UVR-induced development of cutaneous SCC.


Asunto(s)
Carcinoma de Células Escamosas/prevención & control , Proteína Quinasa C-epsilon/genética , Neoplasias Cutáneas/prevención & control , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , Carcinógenos , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa C-epsilon/biosíntesis , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Acetato de Tetradecanoilforbol , Rayos Ultravioleta
15.
Glia ; 64(8): 1285-97, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27145902

RESUMEN

Recent data suggest that ramified microglia fulfil various tasks in the brain. However, to investigate this unique cell type cultured primary microglia are only a poor model. We here describe a method to deplete and repopulate organotypic hippocampal slice cultures (OHSC) with ramified microglia isolated from adult mouse brain creating microglia-replenished OHSC (Mrep-OHSC). Replenished microglia integrate into the tissue and ramify to a degree indistinguishable from their counterparts in the mouse brain. Moreover, wild-type slices replenished with microglia from TNFα-deficient animals provide similar results as OHSC prepared from microglia-specific TNFα-knockout mice (CX3CR1(cre) /TNFα(fl/fl) ). Furthermore, this study demonstrates that replenished microglia in OHSC maintain original functions and properties acquired in vivo. Microglia from ERCC1(Δ/ko) mice, a mouse model of accelerated aging, maintain enhanced Mac2 expression and their activated phenotype after replenishment to wild-type OHSC tissue. Thus, the present study demonstrates that Mrep-OHSC are a unique tool to construct chimeric brain slices allowing studying the function of different phenotypes of in vivo like microglia in a tissue culture setting. GLIA 2016 GLIA 2016;64:1285-1297.


Asunto(s)
Hipocampo/fisiología , Microglía/fisiología , Técnicas de Cultivo de Tejidos , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Galectina 3/metabolismo , Hipocampo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Microscopía Confocal , Neuroprotección/fisiología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
16.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610009

RESUMEN

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Asunto(s)
Colesterol/inmunología , Inmunidad/inmunología , Inflamasomas/inmunología , Interleucina-18/inmunología , Fosfolípidos/inmunología , Saponinas/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Adyuvantes Inmunológicos/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Colesterol/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Combinación de Medicamentos , Humanos , Inmunidad/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Lisosomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosfolípidos/farmacología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/inmunología , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
17.
Acta Pharmacol Sin ; 37(7): 889-97, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27063215

RESUMEN

AIM: MicroRNAs play pivotal roles in regulation of both innate and adaptive immune responses. In the present study, we investigated the effects of microRNA-124 (miR-124) on production of the pro-inflammatory cytokine TNF-α in lipopolysaccharide (LPS)-treated mouse macrophages. METHODS: Mouse macrophage cell line RAW264.7 was stimulated with LPS (100 ng/mL). The levels of miR-124 and TNF-α mRNA were evaluated using q-PCR. ELISA and Western blotting were used to detect TNF-α protein level in cell supernatants and cells, respectively. 3'-UTR luciferase reporter assays were used to analyze the targets of miR-124. For in vivo experiments, mice were injected with LPS (30 mg/kg, ip). RESULTS: LPS stimulation significantly increased the mRNA level of miR-124 in RAW264.7 macrophages in vitro and mice in vivo. In RAW264.7 macrophages, knockdown of miR-124 with miR-124 inhibitor dose-dependently increased LPS-stimulated production of TNF-α protein and prolonged the half-life of TNF-α protein, but did not change TNF-α mRNA levels, whereas overexpression of miR-124 with miR-124 mimic produced the opposite effects. Furthermore, miR-124 was found to directly target two components of deubiquitinating enzymes: ubiquitin-specific proteases (USP) 2 and 14. Knockdown of USP2 or USP14 accelerated protein degradation of TNF-α, and abolished the effect of miR-124 on TNF-α protein stability. CONCLUSION: miR-124, targeting USP2 and USP14, negatively regulates LPS-induced TNF-α production in mouse macrophages, suggesting miR-124 as a new therapeutic target in inflammation-related diseases.


Asunto(s)
Lipopolisacáridos/farmacología , MicroARNs/metabolismo , Estabilidad Proteica , Factor de Necrosis Tumoral alfa/deficiencia , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen , Semivida , Humanos , Macrófagos/metabolismo , Ratones , MicroARNs/sangre , MicroARNs/genética , ARN Interferente Pequeño/farmacología , Transfección , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Ubiquitina Tiolesterasa/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo
18.
Stroke ; 46(8): 2260-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26138121

RESUMEN

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a complex stroke subtype characterized by an initial brain injury, followed by delayed cerebrovascular constriction and ischemia. Current therapeutic strategies nonselectively curtail exacerbated cerebrovascular constriction, which necessarily disrupts the essential and protective process of cerebral blood flow autoregulation. This study identifies a smooth muscle cell autocrine/paracrine signaling network that augments myogenic tone in a murine model of experimental SAH: it links tumor necrosis factor-α (TNFα), the cystic fibrosis transmembrane conductance regulator, and sphingosine-1-phosphate signaling. METHODS: Mouse olfactory cerebral resistance arteries were isolated, cannulated, and pressurized for in vitro vascular reactivity assessments. Cerebral blood flow was measured by speckle flowmetry and magnetic resonance imaging. Standard Western blot, immunohistochemical techniques, and neurobehavioral assessments were also used. RESULTS: We demonstrate that targeting TNFα and sphingosine-1-phosphate signaling in vivo has potential therapeutic application in SAH. Both interventions (1) eliminate the SAH-induced myogenic tone enhancement, but otherwise leave vascular reactivity intact; (2) ameliorate SAH-induced neuronal degeneration and apoptosis; and (3) improve neurobehavioral performance in mice with SAH. Furthermore, TNFα sequestration with etanercept normalizes cerebral perfusion in SAH. CONCLUSIONS: Vascular smooth muscle cell TNFα and sphingosine-1-phosphate signaling significantly enhance cerebral artery tone in SAH; anti-TNFα and anti-sphingosine-1-phosphate treatment may significantly improve clinical outcome.


Asunto(s)
Lisofosfolípidos/biosíntesis , Esfingosina/análogos & derivados , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/fisiopatología , Factor de Necrosis Tumoral alfa/biosíntesis , Vasoconstricción/fisiología , Animales , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/fisiología , Marcación de Gen/métodos , Lisofosfolípidos/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Técnicas de Cultivo de Órganos , Fenilefrina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/biosíntesis , Esfingosina/deficiencia , Hemorragia Subaracnoidea/terapia , Factor de Necrosis Tumoral alfa/deficiencia , Vasoconstricción/efectos de los fármacos , Sistema Vasomotor/efectos de los fármacos , Sistema Vasomotor/fisiología
19.
Kidney Int ; 88(4): 722-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26061548

RESUMEN

Monocyte/macrophage recruitment correlates strongly with the progression of diabetic nephropathy. Tumor necrosis factor-α (TNF-α) is produced by monocytes/macrophages but the direct role of TNF-α and/or macrophage-derived TNF-α in the progression of diabetic nephropathy remains unclear. Here we tested whether inhibition of TNF-α confers kidney protection in diabetic nephropathy via a macrophage-derived TNF-α-dependent pathway. Compared to vehicle-treated mice, blockade of TNF-α with a murine anti-TNF-α antibody conferred kidney protection in Ins2(Akita) mice as indicated by reductions in albuminuria, plasma creatinine, histopathologic changes, kidney macrophage recruitment, and plasma inflammatory cytokine levels at 18 weeks of age. To assess the direct role of macrophage-derived TNF-α in diabetic nephropathy, we generated macrophage-specific TNF-α-deficient mice (CD11b(Cre)/TNF-α(Flox/Flox)). Conditional ablation of TNF-α in macrophages significantly reduced albuminuria, the increase in plasma creatinine and blood urea nitrogen, histopathologic changes, and kidney macrophage recruitment compared to diabetic TNF-α(Flox/Flox) control mice after 12 weeks of streptozotocin-induced diabetes. Thus, production of TNF-α by macrophages plays a major role in diabetic renal injury. Hence, blocking TNF-α could be a novel therapeutic approach for treatment of diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Macrófagos Peritoneales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Albuminuria/prevención & control , Animales , Anticuerpos Neutralizantes/farmacología , Biomarcadores/sangre , Nitrógeno de la Urea Sanguínea , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Quimiotaxis , Creatinina/sangre , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/prevención & control , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/antagonistas & inhibidores , Riñón/efectos de los fármacos , Riñón/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Fenotipo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
20.
J Neuroinflammation ; 12: 125, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-26112704

RESUMEN

BACKGROUND: Tuberculosis (TB) affects one third of the global population, and TB of the central nervous system (CNS-TB) is the most severe form of tuberculosis which often associates with high mortality. The pro-inflammatory cytokine tumour necrosis factor (TNF) plays a critical role in the initial and long-term host immune protection against Mycobacterium tuberculosis (M. tuberculosis) which involves the activation of innate immune cells and structure maintenance of granulomas. However, the contribution of TNF, in particular neuron-derived TNF, in the control of cerebral M. tuberculosis infection and its protective immune responses in the CNS were not clear. METHODS: We generated neuron-specific TNF-deficient (NsTNF(-/-)) mice and compared outcomes of disease against TNF(f/f) control and global TNF(-/-) mice. Mycobacterial burden in brains, lungs and spleens were compared, and cerebral pathology and cellular contributions analysed by microscopy and flow cytometry after M. tuberculosis infection. Activation of innate immune cells was measured by flow cytometry and cell function assessed by cytokine and chemokine quantification using enzyme-linked immunosorbent assay (ELISA). RESULTS: Intracerebral M. tuberculosis infection of TNF(-/-) mice rendered animals highly susceptible, accompanied by uncontrolled bacilli replication and eventual mortality. In contrast, NsTNF(-/-) mice were resistant to infection and presented with a phenotype similar to that in TNF(f/f) control mice. Impaired immunity in TNF(-/-) mice was associated with altered cytokine and chemokine synthesis in the brain and characterised by a reduced number of activated innate immune cells. Brain pathology reflected enhanced inflammation dominated by neutrophil influx. CONCLUSION: Our data show that neuron-derived TNF has a limited role in immune responses, but overall TNF production is necessary for protective immunity against CNS-TB.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Inmunidad Innata/fisiología , Mycobacterium tuberculosis/fisiología , Neuronas/microbiología , Neuronas/patología , Tuberculosis del Sistema Nervioso Central/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Replicación Viral/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/microbiología , Encéfalo/patología , Proliferación Celular/fisiología , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/microbiología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Microglía/microbiología , Microglía/patología , Tuberculosis del Sistema Nervioso Central/patología , Tuberculosis del Sistema Nervioso Central/fisiopatología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA