Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Immunol ; 187(4): 1702-12, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21734073

RESUMEN

TNF has been implicated in the pathogenesis of type 1 diabetes. When administered early in life, TNF accelerates and increases diabetes in NOD mice. However, when administered late, TNF decreases diabetes incidence and delays onset. TNFR1-deficient NOD mice were fully protected from diabetes and only showed mild peri-insulitis. To further dissect how TNFR1 deficiency affects type 1 diabetes, these mice were crossed to ß cell-specific, highly diabetogenic TCR transgenic I-A(g7)-restricted NOD4.1 mice and Kd-restricted NOD8.3 mice. TNFR1-deficient NOD4.1 and NOD8.3 mice were protected from diabetes and had significantly less insulitis compared with wild type NOD4.1 and NOD8.3 controls. Diabetic NOD4.1 mice rejected TNFR1-deficient islet grafts as efficiently as control islets, confirming that TNFR1 signaling is not directly required for ß cell destruction. Flow cytometric analysis showed a significant increase in the number of CD4(+)CD25(+)Foxp3(+) T regulatory cells in TNFR1-deficient mice. TNFR1-deficient T regulatory cells were functionally better at suppressing effector cells than were wild type T regulatory cells both in vitro and in vivo. This study suggests that blocking TNF signaling may be beneficial in increasing the function of T regulatory cells and suppression of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Diabetes Mellitus Tipo 1/genética , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética , Trasplante Homólogo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología
2.
Proc Natl Acad Sci U S A ; 106(47): 20057-62, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19897719

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid-beta (Abeta)-containing plaques, neurofibrillary tangles, and neuron and synapse loss. Tangle formation has been reproduced in P301L tau transgenic pR5 mice, whereas APP(sw)PS2(N141I) double-transgenic APP152 mice develop Abeta plaques. Cross-breeding generates triple transgenic ((triple)AD) mice that combine both pathologies in one model. To determine functional consequences of the combined Abeta and tau pathologies, we performed a proteomic analysis followed by functional validation. Specifically, we obtained vesicular preparations from (triple)AD mice, the parental strains, and nontransgenic mice, followed by the quantitative mass-tag labeling proteomic technique iTRAQ and mass spectrometry. Within 1,275 quantified proteins, we found a massive deregulation of 24 proteins, of which one-third were mitochondrial proteins mainly related to complexes I and IV of the oxidative phosphorylation system (OXPHOS). Notably, deregulation of complex I was tau dependent, whereas deregulation of complex IV was Abeta dependent, both at the protein and activity levels. Synergistic effects of Abeta and tau were evident in 8-month-old (triple)AD mice as only they showed a reduction of the mitochondrial membrane potential at this early age. At the age of 12 months, the strongest defects on OXPHOS, synthesis of ATP, and reactive oxygen species were exhibited in the (triple)AD mice, again emphasizing synergistic, age-associated effects of Abeta and tau in perishing mitochondria. Our study establishes a molecular link between Abeta and tau protein in AD pathology in vivo, illustrating the potential of quantitative proteomics.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Fosforilación Oxidativa , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Humanos , Espectrometría de Masas/métodos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas tau/genética
3.
J Neurosci ; 29(5): 1319-30, 2009 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19193879

RESUMEN

Microglia and infiltrating leukocytes are considered major producers of tumor necrosis factor (TNF), which is a crucial player in cerebral ischemia and brain inflammation. We have identified a neuroprotective role for microglial-derived TNF in cerebral ischemia in mice. We show that cortical infarction and behavioral deficit are significantly exacerbated in TNF-knock-out (KO) mice compared with wild-type mice. By using in situ hybridization, immunohistochemistry, and green fluorescent protein bone marrow (BM)-chimeric mice, TNF was shown to be produced by microglia and infiltrating leukocytes. Additional analysis demonstrating that BM-chimeric TNF-KO mice grafted with wild-type BM cells developed larger infarcts than BM-chimeric wild-type mice grafted with TNF-KO BM cells provided evidence that the neuroprotective effect of TNF was attributable to microglial- not leukocyte-derived TNF. In addition, observation of increased infarction in TNF-p55 receptor (TNF-p55R)-KO mice compared with TNF-p75R and wild-type mice suggested that microglial-derived TNF exerts neuroprotective effects through TNF-p55R. We finally report that TNF deficiency is associated with reduced microglial population size and Toll-like receptor 2 expression in unmanipulated brain, which might also influence the neuronal response to injury. Our results identify microglia and microglial-derived TNF as playing a key role in determining the survival of endangered neurons in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Microglía/metabolismo , Neuronas/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Microglía/fisiología , Neuronas/patología , Factor de Necrosis Tumoral alfa/fisiología
4.
J Neurosci ; 28(19): 5072-81, 2008 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-18463260

RESUMEN

To provide a tool to investigate the mechanisms inducing and maintaining cancer-related pain and hyperalgesia, a soft tissue tumor/metastasis model was developed that is applicable in C57BL/6J wild-type and transgenic mice. We show that the experimental tumor-induced heat hyperalgesia and nociceptor sensitization were prevented by systemic treatment with the tumor necrosis factor alpha (TNFalpha) antagonist etanercept. In naive mice, exogenous TNFalpha evoked heat hyperalgesia in vivo and sensitized nociceptive nerve fibers to heat in vitro. TNFalpha enhanced the expression of the nociceptor-specific heat transducer ion channel transient receptor potential vanilloid 1 (TRPV1) and increased the amplitudes of capsaicin and heat-activated ionic currents via p38/MAP (mitogen-activated protein) kinase and PKC (protein kinase C). Deletion of the tumor necrosis factor receptor type 2 (TNFR2) gene attenuated heat hyperalgesia and prevented TRPV1 upregulation in tumor-bearing mice, whereas TNFR1 gene deletion played a minor role. We propose endogenous TNFalpha as a key player in cancer-related heat hyperalgesia and nociceptor sensitization that generates TRPV1 upregulation and sensitization via TNFR2.


Asunto(s)
Carcinoma/complicaciones , Carcinoma/metabolismo , Hiperalgesia/etiología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Capsaicina/farmacología , Células Cultivadas , Etanercept , Eliminación de Gen , Miembro Posterior , Calor , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Inmunoglobulina G/farmacología , Ratones , Trasplante de Neoplasias , Neuronas Aferentes/efectos de los fármacos , Nociceptores/efectos de los fármacos , Nociceptores/fisiopatología , Técnicas de Placa-Clamp , Receptores del Factor de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Canales Catiónicos TRPV/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
5.
J Pharmacol Exp Ther ; 324(3): 948-56, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18083911

RESUMEN

The recent identification of the trace amine-associated receptor (TAAR)1 provides an opportunity to dissociate the effects of trace amines on the dopamine transporter from receptor-mediated effects. To separate both effects on a physiological level, a Taar1 knockout mouse line was generated. Taar1 knockout mice display increased sensitivity to amphetamine as revealed by enhanced amphetamine-triggered increases in locomotor activity and augmented striatal release of dopamine compared with wild-type animals. Under baseline conditions, locomotion and extracellular striatal dopamine levels were similar between Taar1 knockout and wild-type mice. Electrophysiological recordings revealed an elevated spontaneous firing rate of dopaminergic neurons in the ventral tegmental area of Taar1 knock-out mice. The endogenous TAAR1 agonist p-tyramine specifically decreased the spike frequency of these neurons in wild-type but not in Taar1 knockout mice, consistent with the prominent expression of Taar1 in the ventral tegmental area. Taken together, the data reveal TAAR1 as regulator of dopaminergic neurotransmission.


Asunto(s)
Antagonistas de Dopamina/metabolismo , Dopamina/metabolismo , Actividad Motora/fisiología , Receptores Acoplados a Proteínas G/biosíntesis , Área Tegmental Ventral/fisiología , Anfetamina/farmacología , Animales , Dopamina/genética , Dopamina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
6.
J Neurosci ; 22(15): 6713-23, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12151550

RESUMEN

NMDA receptor hypofunction has been implicated in the pathophysiology of schizophrenia, and pharmacological and genetic approaches have been used to model such dysfunction. We previously have described two mouse lines carrying point mutations in the NMDA receptor glycine binding site, Grin1(D481N) and Grin1(K483Q), which exhibit 5- and 86-fold reductions in receptor glycine affinity, respectively. Grin1(D481N) animals exhibit a relatively mild phenotype compatible with a moderate reduction in NMDA receptor function, whereas Grin1(K483Q) animals die shortly after birth. In this study we have characterized compound heterozygote Grin1(D481N/K483Q) mice, which are viable and exhibited biphasic NMDA receptor glycine affinities compatible with the presence of each of the two mutated alleles. Grin1(D481N/K483Q) mice exhibited a marked NMDA receptor hypofunction revealed by deficits in hippocampal long-term potentiation, which were rescued by the glycine site agonist d-serine, which also facilitated NMDA synaptic currents in mutant, but not in wild-type, mice. Analysis of striatal monoamine levels revealed an apparent dopaminergic and serotonergic hyperfunction. Behaviorally, Grin1(D481N/K483Q) mice were insensitive to acute dizocilpine pretreatment and exhibited increased startle response but normal prepulse inhibition. Most strikingly, mutant mice exhibited a sustained, nonhabituating hyperactivity and increased stereotyped behavior that were resistant to suppression by antipsychotics and the benzodiazepine site agonist Zolpidem. They also displayed a disruption of nest building behavior and were unable to perform a cued learning paradigm in the Morris water maze. We speculate that the severity of NMDA receptor hypofunction in these mice may account for their profound behavioral phenotype and insensitivity to antipsychotics.


Asunto(s)
Resistencia a Medicamentos/genética , Glicina/metabolismo , Hipercinesia/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Sitios de Unión/genética , Unión Competitiva/efectos de los fármacos , Unión Competitiva/genética , Aminas Biogénicas/metabolismo , Cuerpo Estriado/química , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Agonistas del GABA/farmacología , Marcación de Gen , Ácido Glutámico/farmacocinética , Glicina/agonistas , Glicina/farmacocinética , Heterocigoto , Hipocampo/fisiopatología , Hipercinesia/fisiopatología , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Ratones , Ratones Mutantes Neurológicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Fenotipo , Mutación Puntual , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Serina/análogos & derivados , Serina/farmacología , Estereoisomerismo , Conducta Estereotipada/efectos de los fármacos
7.
J Neurosci ; 23(26): 8989-9003, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14523101

RESUMEN

Transgenic mice, expressing mutant beta-amyloid precursor proteins (betaAPPs), have lead to a better understanding of the pathophysiological processes in Alzheimer's disease (AD). In many of these models, however, the temporal development of cognitive decline and the relationship to Abeta deposition and inflammation are unclear. We now report a novel transgenic mouse line, PS2APP (PS2N141I x APPswe), which develops a severe cerebral amyloidosis in discrete brain regions, and present a cross-sectional analysis of these mice at 4, 8, 12, and 16 months of age. Each age cohort was investigated for changes in behavior, electrophysiology of synapse efficacy, ELISA-determined Abeta load, histopathology, and in immunoelectron microscopy. Cognitive deficits were first observed at 8 months when Abeta deposits and inflammation were restricted to discrete brain regions, namely the subiculum and frontolateral (motor and orbital) cortex. As early as 5 months, electron microscopy revealed the presence, in these regions, of pre-plaque, immunogold-labeled extracellular fibrillar Abeta. At the same age, increased levels of insoluble Abeta were detected by ELISA, with Abeta1-40 levels exceeding those of Abeta1-42. Further cognitive decline occurred in an age-related manner, and this was accompanied by the spread of amyloidosis to ultimately affect not only neo- and limbic cortices, but also thalamic and pontine nuclei. Dentate gyrus post-tetanic potentiation was significantly attenuated at 17 months, and there were also significant differences in paired-pulse parameters. This systematic cross-sectional study of the behavioral and pathological changes in the PS2APP mouse indicates that it develops age-related cognitive decline associated with severe amyloidosis and inflammation in discrete brain regions and therefore is suitable for testing a range of potential symptomatic and disease-modifying therapies for AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/biosíntesis , Amiloidosis/fisiopatología , Encéfalo/metabolismo , Trastornos del Conocimiento/fisiopatología , Proteínas de la Membrana/biosíntesis , Factores de Edad , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/complicaciones , Amiloidosis/patología , Animales , Conducta Animal , Encéfalo/patología , Química Encefálica , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Estudios Transversales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipocampo/fisiopatología , Humanos , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microscopía Inmunoelectrónica , Mutación , Plasticidad Neuronal/genética , Presenilina-2 , Transmisión Sináptica/genética
8.
Neurosci Lett ; 373(1): 79-84, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15555781

RESUMEN

The occupation of the glycine binding-site is a prerequisite for NMDA receptor activation by glutamate. To analyze the regulation of NMDA receptor function by the glycine transporter 1 (GlyT1), we generated heterozygous constitutive GlyT1 knockout mice (GlyT1tm1.1(+/-)). These animals were fully viable. Using a newly generated antibody, the pattern of GlyT1 expression in brain was found to be unaltered in the mutants while the level of expression was strongly reduced in all brain regions, as shown immunohistochemically. In hippocampal slices the ratio of the peak amplitude of NMDA and AMPA receptor evoked excitatory postsynaptic currents (EPSCs), recorded in CA1 pyramidal cells, was significantly enhanced by 36% in Glyt1tm1.1(+/-) compared to wild-type slices. The frequency and amplitude of AMPA miniature events in Glyt1tm1.1(+/-) mice were indistinguishable from those recorded in wild type. These results provide proof that the NMDA receptor function is enhanced by a reduction of GlyT1 expression. Thus, GlyT1 function is a controlling factor for an enhancement of the NMDA receptor response. These findings are of relevance for the development of GlyT1 inhibitory drugs.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Encéfalo/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/efectos de los fármacos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Southern Blotting , Western Blotting , Encéfalo/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Glicina/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática , Inmunohistoquímica , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos
9.
Am J Pathol ; 170(3): 990-1002, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322383

RESUMEN

Listeria monocytogenes may infect the central nervous system and several peripheral organs. To explore the function of IL-1 receptor type 1 (IL-1R1) in cerebral versus systemic listeriosis, IL-1R1(-/-) and wild-type mice were infected either intracerebrally or intraperitoneally with L. monocytogenes. After intracerebral infection with various numbers of attenuated Listeria, IL-1R1(-/-) mice succumbed due to an insufficient control of intracerebral Listeria, whereas all wild-type mice survived, efficiently restricting growth of Listeria. IL-1R1(-/-) mice recruited increased numbers of leukocytes, especially granulocytes, to the brain compared with wild-type mice. In contrast, both IL-1R1(-/-) and wild-type mice survived a primary and secondary intraperitoneal infection with Listeria without differences in the hepatic bacterial load. In addition, both strains developed similar frequencies of Listeria-specific CD4 and CD8 T cells after primary and secondary intraperitoneal infection. However, an intraperitoneal immunization before intracerebral challenge infection neither protected IL-1R1(-/-) mice from death nor reduced the intracerebral bacterial load, although numbers of intracerebral Listeria-specific CD4 and CD8 T cells and levels of inducible nitric oxide synthase, tumor necrosis factor, and interferon-gamma mRNA were identical in IL-1R1(-/-) and wild-type mice. Collectively, these findings illustrate a crucial role of IL-1R1 in cerebral but not systemic listeriosis.


Asunto(s)
Listeriosis/inmunología , Listeriosis/metabolismo , Meningitis por Listeria/inmunología , Meningitis por Listeria/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/análisis , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Interleucina-1alfa/análisis , Interleucina-1alfa/metabolismo , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Listeriosis/patología , Masculino , Meningitis por Listeria/patología , Ratones , Ratones Mutantes , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Immunol ; 179(9): 5644-8, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17947634

RESUMEN

Sphingosine kinase (Sphk) phosphorylates sphingosine into sphingosine-1-phosphate (S1P), but its recently identified isoform Sphk2 has been suggested to have distinct subcellular localization and substrate specificity. We demonstrate here that, surprisingly, Sphk2(-/-) CD4(+) T cells exhibit a hyperactivated phenotype with significantly enhanced proliferation and cytokine secretion in response to IL-2 as well as reduced sensitivity to regulatory T cell-mediated suppression in vitro, apparently independent of effects upon S1P. Such findings appear to reflect a requirement for Sphk2 to suppress IL-2 signaling because, in Sphk2(-/-) CD4(+) T cells, IL-2 induced abnormally accentuated STAT5 phosphorylation and small interfering RNA knockdown of STAT5 abrogated their hyperactive phenotype. This pathway physiologically modulates autoinflammatory responses, because Sphk2(-/-) T cells induced more rapid and robust inflammatory bowel disease in scid recipients. Thus, Sphk2 regulates IL-2 pathways in T cells, and the modulation of Sphk2 activity may be of therapeutic utility in inflammatory and/or infectious diseases.


Asunto(s)
Autoinmunidad/inmunología , Interleucina-2/inmunología , Interleucina-2/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal/inmunología , Animales , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Lisofosfolípidos/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfocitos T/enzimología , Linfocitos T/inmunología
11.
J Virol ; 80(13): 6339-44, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16775322

RESUMEN

To understand the correlates of protective immunity against primary variola virus infection in humans, we have used the well-characterized mousepox model. This is an excellent surrogate small-animal model for smallpox in which the disease is caused by infection with the closely related orthopoxvirus, ectromelia virus. Similarities between the two infections include virus replication and transmission, aspects of pathology, and development of pock lesions. Previous studies using ectromelia virus have established critical roles for cytokines and effector functions of CD8 T cells in the control of acute stages of poxvirus infection. Here, we have used mice deficient in B cells to demonstrate that B-cell function is also obligatory for complete virus clearance and recovery of the host. In the absence of B cells, virus persists and the host succumbs to infection, despite the generation of CD8 T-cell responses. Intriguingly, transfer of naive B cells or ectromelia virus-immune serum to B-cell-deficient mice with established infection allowed these animals to clear virus and fully recover. In contrast, transfer of ectromelia virus-immune CD8 T cells was ineffective. Our data show that mice deficient in CD8 T-cell function die early in infection, whereas those deficient in B cells or antibody production die much later, indicating that B-cell function becomes critical after the effector phase of the CD8 T-cell response to infection subsides. Strikingly, our results show that antibody prevents virus from seeding the skin and forming pock lesions, which are important for virus transmission between hosts.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/transmisión , Traslado Adoptivo , Animales , Anticuerpos Antivirales/inmunología , Linfocitos B/trasplante , Linfocitos T CD8-positivos/trasplante , Modelos Animales de Enfermedad , Ectromelia Infecciosa/patología , Femenino , Humanos , Inmunidad Celular/inmunología , Depleción Linfocítica , Ratones , Ratones Noqueados , Piel/inmunología , Piel/patología , Piel/virología , Viruela/inmunología , Viruela/patología , Viruela/transmisión , Virus de la Viruela/inmunología , Replicación Viral/inmunología
12.
Infect Immun ; 70(10): 5857-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12228317

RESUMEN

Using tumor necrosis factor receptor type 2 (TNFR2)-deficient mice and generating bone marrow chimeras which express TNFR2 on either hematopoietic or nonhematopoietic cells, we demonstrated the requirement for TNFR2 expression on tissue cells to induce lethal cerebral malaria. Thus, TNFR2 on the brain vasculature mediates tumor necrosis factor-induced neurovascular lesions in experimental cerebral malaria.


Asunto(s)
Antígenos CD/fisiología , Vasos Sanguíneos/inmunología , Malaria Cerebral/etiología , Plasmodium berghei , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Antígenos CD/genética , Plaquetas/patología , Trasplante de Médula Ósea , Circulación Cerebrovascular/inmunología , Quimera , Endotelio Vascular/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parasitemia/etiología , Parasitemia/inmunología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral
13.
Mol Cell Neurosci ; 24(2): 442-50, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14572465

RESUMEN

The gamma2 subunit of GABA(A) receptor chloride channels is required for normal channel function and for postsynaptic clustering of these receptors during synaptogenesis. In addition, GABA(A) receptor function is thought to contribute to normal postnatal maturation of neurons. Loss of postsynaptic GABA(A) receptors in gamma2-deficient neurons might therefore reflect a deficit in maturation of neurons due to the reduced channel function. Here, we have used the Cre-loxP strategy to examine the clustering function of the gamma2 subunit at mature synapses. Deletion of the gamma2 subunit in the third postnatal week resulted in loss of benzodiazepine-binding sites and parallel loss of punctate immunoreactivity for postsynaptic GABA(A) receptors and gephyrin. Thus, the gamma2 subunit contributes to postsynaptic localization of GABA(A) receptors and gephyrin by a mechanism that is operant in mature neurons and not limited to immature neurons, most likely through interaction with proteins involved in trafficking of synaptic GABA(A) receptors.


Asunto(s)
Encéfalo/metabolismo , Receptores de GABA-A/fisiología , Sinapsis/genética , Sinapsis/metabolismo , Animales , Sitios de Unión/fisiología , Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de GABA-A/biosíntesis , Receptores de GABA-A/deficiencia , Receptores de GABA-A/genética
14.
J Biol Chem ; 279(32): 34032-7, 2004 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-15163665

RESUMEN

ATP binding cassette transporter A1 (ABCA1) is involved in regulation of intracellular lipid trafficking and export of cholesterol from cells to high density lipoproteins. ABCA1 defects cause Tangier disease, a disorder characterized by absence of high density lipoprotein and thrombocytopenia. In the present study we have demonstrated that ABCA1 is expressed in human platelets and that fibrinogen binding and CD62 surface expression in response to collagen and low concentrations of thrombin, but not to ADP, are defective in platelets from Tangier patients and ABCA1-deficient animals. The expression of platelet membrane receptors such as GPVI, alpha2beta1 integrin, and GPIIb/IIIa, the collagen-induced changes in phosphatidylserine and cholesterol distribution, and the collagen-induced signal transduction examined by phosphorylation of LAT and p72syk and by intracellular Ca2+ mobilization were unaltered in Tangier platelets. The electron microscopy of Tangier platelets revealed reduced numbers of dense bodies and the presence of giant granules typically encountered in platelets from Chediak-Higashi syndrome. Further studies demonstrated impaired release of dense body content in platelets from Tangier patients and ABCA1-deficient animals. In addition, Tangier platelets were characterized by defective surface exposure of dense body and lysosomal markers (CD63, LAMP-1, LAMP-2, CD68) during collagen- and thrombin-induced stimulation and by abnormally high lysosomal pH. We conclude that intact ABCA1 function is necessary for proper maturation of dense bodies in platelets. The impaired release of the content of dense bodies may explain the defective activation of Tangier platelets by collagen and low concentrations of thrombin, but not by ADP.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Activación Plaquetaria/fisiología , Enfermedad de Tangier/sangre , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/sangre , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Difosfato/farmacología , Anciano , Animales , Antígenos CD/sangre , Antígenos de Diferenciación Mielomonocítica/sangre , Plaquetas/química , Plaquetas/fisiología , Plaquetas/ultraestructura , Moléculas de Adhesión Celular/sangre , Colágeno/sangre , Colágeno/farmacología , Gránulos Citoplasmáticos/fisiología , Gránulos Citoplasmáticos/ultraestructura , Fibrinógeno/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Proteínas de Membrana de los Lisosomas , Lisosomas/química , Lisosomas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Persona de Mediana Edad , Fosfatidilserinas/sangre , Activación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria , Tetraspanina 30 , Trombina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA