Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(21): 8867-72, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21555578

RESUMO

Estrogen has well-documented neuroprotective effects in a variety of clinical and experimental disorders of the CNS, including autoimmune inflammation, traumatic injury, stroke, and neurodegenerative diseases. The beneficial effects of estrogens in CNS disorders include mitigation of clinical symptoms, as well as attenuation of histopathological signs of neurodegeneration and inflammation. The cellular mechanisms that underlie these CNS effects of estrogens are uncertain, because a number of different cell types express estrogen receptors in the peripheral immune system and the CNS. Here, we investigated the potential roles of two endogenous CNS cell types in estrogen-mediated neuroprotection. We selectively deleted estrogen receptor-α (ERα) from either neurons or astrocytes using well-characterized Cre-loxP systems for conditional gene knockout in mice, and studied the effects of these conditional gene deletions on ERα ligand-mediated neuroprotective effects in a well-characterized model of adoptive experimental autoimmune encephalomyelitis (EAE). We found that the pronounced and significant neuroprotective effects of systemic treatment with ERα ligand on clinical function, CNS inflammation, and axonal loss during EAE were completely prevented by conditional deletion of ERα from astrocytes, whereas conditional deletion of ERα from neurons had no significant effect. These findings show that signaling through ERα in astrocytes, but not through ERα in neurons, is essential for the beneficial effects of ERα ligand in EAE. Our findings reveal a unique cellular mechanism for estrogen-mediated CNS neuroprotective effects by signaling through astrocytes, and have implications for understanding the pathophysiology of sex hormone effects in diverse CNS disorders.


Assuntos
Encefalomielite Autoimune Experimental/patologia , Receptor alfa de Estrogênio/fisiologia , Fármacos Neuroprotetores/farmacologia , Animais , Astrócitos/patologia , Células Cultivadas , Receptor alfa de Estrogênio/deficiência , Inflamação/prevenção & controle , Ligantes , Camundongos , Camundongos Knockout , Doenças Neurodegenerativas/prevenção & controle , Neurônios/patologia
2.
J Neurosci ; 32(36): 12312-24, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956822

RESUMO

Over 50% of multiple sclerosis (MS) patients experience cognitive deficits, and hippocampal-dependent memory impairment has been reported in >30% of these patients. While postmortem pathology studies and in vivo magnetic resonance imaging demonstrate that the hippocampus is targeted in MS, the neuropathology underlying hippocampal dysfunction remains unknown. Furthermore, there are no treatments available to date to effectively prevent neurodegeneration and associated cognitive dysfunction in MS. We have recently demonstrated that the hippocampus is also targeted in experimental autoimmune encephalomyelitis (EAE), the most widely used animal model of MS. The objective of this study was to assess whether a candidate treatment (testosterone) could prevent hippocampal synaptic dysfunction and underlying pathology when administered in either a preventative or a therapeutic (postdisease induction) manner. Electrophysiological studies revealed impairments in basal excitatory synaptic transmission that involved both AMPA receptor-mediated changes in synaptic currents, and faster decay rates of NMDA receptor-mediated currents in mice with EAE. Neuropathology revealed atrophy of the pyramidal and dendritic layers of hippocampal CA1, decreased presynaptic (Synapsin-1) and postsynaptic (postsynaptic density 95; PSD-95) staining, diffuse demyelination, and microglial activation. Testosterone treatment administered either before or after disease induction restores excitatory synaptic transmission as well as presynaptic and postsynaptic protein levels within the hippocampus. Furthermore, cross-modality correlations demonstrate that fluctuations in EPSPs are significantly correlated to changes in postsynaptic protein levels and suggest that PSD-95 is a neuropathological substrate to impaired synaptic transmission in the hippocampus during EAE. This is the first report demonstrating that testosterone is a viable therapeutic treatment option that can restore both hippocampal function and disease-associated pathology that occur during autoimmune disease.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Guanilato Quinases/fisiologia , Hipocampo/metabolismo , Proteínas de Membrana/fisiologia , Polirradiculoneuropatia/tratamento farmacológico , Transmissão Sináptica/fisiologia , Testosterona/administração & dosagem , Animais , Proteína 4 Homóloga a Disks-Large , Implantes de Medicamento , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polirradiculoneuropatia/metabolismo , Polirradiculoneuropatia/patologia , Distribuição Aleatória , Transmissão Sináptica/efeitos dos fármacos
3.
Neuroimage ; 60(1): 95-104, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22182769

RESUMO

There are strong correlations between cortical atrophy observed by MRI and clinical disability and disease duration in multiple sclerosis (MS). The objective of this study was to evaluate the progression of cortical atrophy over time in vivo in experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model for MS. Volumetric changes in brains of EAE mice and matched healthy controls were quantified by collecting high-resolution T2-weighted magnetic resonance images in vivo and labeling anatomical structures on the images. In vivo scanning permitted us to evaluate brain structure volumes in individual animals over time and we observed that though brain atrophy progressed differently in each individual animal, all mice with EAE demonstrated significant atrophy in whole brain, cerebral cortex, and whole cerebellum compared to normal controls. Furthermore, we found a strong correlation between cerebellar atrophy and cumulative disease score in mice with EAE. Ex vivo MRI showed a significant decrease in brain and cerebellar volume and a trend that did not reach significance in cerebral cortex volume in mice with EAE compared to controls. Cross modality correlations revealed a significant association between neuronal loss on neuropathology and in vivo atrophy of the cerebral cortex by neuroimaging. These results demonstrate that longitudinal in vivo imaging is more sensitive to changes that occur in neurodegenerative disease models than cross-sectional ex vivo imaging. This is the first report of progressive cortical atrophy in vivo in a mouse model of MS.


Assuntos
Cerebelo/patologia , Córtex Cerebral/patologia , Encefalomielite Autoimune Experimental/patologia , Imageamento por Ressonância Magnética , Animais , Atrofia , Camundongos
4.
Eur J Immunol ; 41(1): 140-50, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21182085

RESUMO

Estrogens act upon nuclear estrogen receptors (ER) to ameliorate cell-mediated autoimmune disease. As most immunomodulatory effects of estrogens in EAE have been attributed to the function of ER-α, we previously demonstrated that ER-ß ligand treatment reduced disease severity without affecting peripheral cytokine production or levels of CNS inflammation, suggesting a direct neuroprotective effect; however, the effect of ER-ß treatment on the function of immune cells within the target organ remained unknown. Here, we used adoptive transfer studies to show that ER-ß ligand treatment was protective in the effector, but not the induction phase of EAE, as shown by decreased clinical disease severity with the preservation of axons and myelin in spinal cords. The analysis of the immune cell infiltrates in the CNS revealed that while ER-ß ligand treatment did not reduce overall levels of CNS inflammation, there was a decrease in the DC percentage, and these CNS DC had decreased TNF-α production. Finally, experiments using DC deficient in ER-ß revealed that the expression of ER-ß on DC was essential for protective effects of ER-ß ligand treatment in EAE. Our results demonstrate for the first time an effect of ER-ß ligand treatment in vivo on DC in the target organ of a prototypic cell-mediated autoimmune disease.


Assuntos
Transferência Adotiva , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/imunologia , Animais , Axônios/efeitos dos fármacos , Axônios/imunologia , Células Dendríticas/efeitos dos fármacos , Encefalomielite Autoimune Experimental/imunologia , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/imunologia , Nitrilas/farmacologia , Propionatos/farmacologia , Índice de Gravidade de Doença , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/imunologia
5.
J Neurosci Res ; 90(7): 1310-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22411609

RESUMO

Gray matter atrophy is an important correlate to clinical disability in multiple sclerosis (MS), and many treatment trials include atrophy as an outcome measure. Atrophy has been shown to occur in experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model of MS. The clinical severity of EAE is reduced in estrogen-reated mice, but it remains unknown whether estrogen treatment can reduce gray matter atrophy in EAE. In this study, mice with EAE were treated with either estrogen receptor (ER)-α ligand or ER-ß ligand, and diffusion tensor images (DTI) were collected and neuropathology was performed. DTI showed atrophy in the cerebellar gray matter of vehicle-treated EAE mice compared with healthy controls but not in ER-α or ER-ß ligand-treated EAE mice. Neuropathology demonstrated that Purkinje cell numbers were decreased in vehicle-treated EAE mice, whereas neither ER ligand-treated EAE groups showed a decrease. This is the first report of a neuroprotective therapy in EAE that unambiguously prevents gray matter atrophy while sparing a major neuronal cell type. Fractional anisotropy (FA) in the cerebellar white matter was decreased in vehicle- and ER-ß ligand-treated but not in ER-α ligand-treated EAE mice. Inflammatory cell infiltration was increased in vehicle- and ER-ß ligand-treated but not in ER-α ligand-treated EAE mice. Myelin staining was decreased in vehicle-treated EAE mice and was spared in both ER ligand-treated groups. This is consistent with decreased FA as a potential biomarker for inflammation rather than myelination or axonal damage in the cerebellum in EAE.


Assuntos
Encéfalo/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Estrogênios/farmacologia , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Atrofia , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Estrogênios/uso terapêutico , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Degeneração Neural/patologia , Fármacos Neuroprotetores/uso terapêutico , Resultado do Tratamento
6.
Nat Neurosci ; 10(11): 1398-400, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17952068

RESUMO

Sex differences in brain function and behavior are regularly attributed to gonadal hormones. Some brain sexual dimorphisms, however, are direct actions of sex chromosome genes that are not mediated by gonadal hormones. We used mice in which sex chromosome complement (XX versus XY) and gonadal sex (ovaries versus testes) were independent, and found that XX mice showed faster food-reinforced instrumental habit formation than XY mice, regardless of gonadal phenotype.


Assuntos
Transtornos do Desenvolvimento Sexual , Genes sry/fisiologia , Hábitos , Caracteres Sexuais , Cromossomos Sexuais/fisiologia , Animais , Antimaníacos/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Comportamento Animal , Feminino , Genes sry/genética , Cloreto de Lítio/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Cromossomos Sexuais/genética , Processos de Determinação Sexual , Cromossomo X , Cromossomo Y
7.
Oncotarget ; 6(17): 15137-49, 2015 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-25883222

RESUMO

Androgen Receptor (AR) is the male hormone receptor and a nuclear transcription factor which plays a central role in the growth of normal and malignant prostate gland. Our earlier studies defined a mechanistic model for male hormone dependent regulation of AR protein levels in prostate cancer (CaP) cells through a negative feed-back loop between AR and PMEPA1, an androgen induced NEDD4 E3 ubiquitin ligase binding protein. This report focuses on the impact of PMEPA1 silencing on CaP biology. PMEPA1 knockdown accelerated the growth of CaP tumor cells in athymic nude mice. In cell culture models knockdown of PMEPA1 resulted in resistance to AR inhibitors enzalutamide and bicalutamide. While, AR protein down regulation by NEDD4 was PMEPA1 dependent, we also noted a PMEPA1 independent downregulation of PTEN by NEDD4. In a subset of human CaP, decreased PMEPA1 mRNA expression significantly correlated with increased levels of AR transcription target PSA, as a surrogate for elevated AR. This study highlights that silencing of PMEPA1 accelerates the growth of CaP cells through AR, NEDD4 and PTEN. Thus, the therapeutic restoration of PMEPA1 represents a promising complementary strategy correcting for AR and PTEN defects in CaP. Statement of significance: Here we define that silencing of PMEPA1 facilitates the growth of CaP cells and modulates AR through NEDD4 and PTEN. The restoration of PMEPA1 represents a promising complementary therapeutic strategy correcting for AR and PTEN defects.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas de Membrana/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/metabolismo , Interferência de RNA , Receptores Androgênicos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Animais , Benzamidas , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/genética , Camundongos Nus , Ubiquitina-Proteína Ligases Nedd4 , Nitrilas/farmacologia , PTEN Fosfo-Hidrolase/genética , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/genética , Compostos de Tosil/farmacologia , Transplante Heterólogo , Carga Tumoral/genética , Ubiquitina-Proteína Ligases/genética
8.
J Clin Endocrinol Metab ; 97(8): E1402-10, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22659246

RESUMO

CONTEXT: Glucocorticoids (GC) are powerful endogenous and therapeutic modulators of inflammation and play a critical role for controlling autoimmunity. GC resistance can be seen in patients with cell-mediated autoimmune disorders, but it is unknown whether this represents a stable trait or a state. OBJECTIVE: The objective of the study was to determine whether GC resistance of T cell responses is dynamically regulated in experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS). DESIGN: This was a translational observational study. PATIENTS AND ANIMALS: EAE was induced in C57BL/6 mice. A cross-sectional sample of 25 patients with relapsing-remitting MS was included as well as four MS patients during pregnancy and postpartum. MAIN OUTCOME MEASURES: Outcome measures included GC sensitivity of T cell proliferation and GC-mediated apoptosis. RESULTS: GC resistance was seen in both autoantigen-specific and nonspecific responses of T cells obtained from mice with EAE. GC resistance preceded clinical symptoms and central nervous system infiltration of immune cells. T cells obtained during EAE were resistant to GC-induced apoptosis, and this was linked to down-regulation of GC receptor-α expression. GC resistance in T cells was also seen in MS patients with radiological evidence for ongoing inflammation. GC resistance was absent in the MS patients during pregnancy, when relapse risk is decreased, but recurred postpartum, a time of increased relapse risk. CONCLUSIONS: These data demonstrate that GC resistance during autoimmune neuroinflammation is dynamically regulated. This has implications for the timing of steroid treatments and provides a putative pathway to explain the observed association between psychological stress and exacerbation of autoimmune diseases.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Glucocorticoides/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Autoantígenos/imunologia , Estudos Transversais , Resistência a Medicamentos , Encefalomielite Autoimune Experimental/imunologia , Feminino , Glicoproteínas/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/imunologia , Receptores de Glucocorticoides/genética , Linfócitos T/imunologia
9.
J Pain ; 9(10): 962-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18635401

RESUMO

UNLABELLED: In animal studies of nociception, females are often more sensitive to painful stimuli, whereas males are often more sensitive to analgesia induced by mu-agonists. Sex differences are found even at birth, and in adulthood are likely caused, at least in part, by differences in levels of gonadal hormones. In this report, we investigate nociception and analgesia in neonatal mice and assess the contribution of the direct action of sex chromosome genes in hotplate and tail withdrawal tests. We used the 4 core genotypes mouse model, in which gonadal sex is independent of the complement of sex chromosomes (XX vs XY). Mice were tested at baseline and then injected with mu-opioid agonist morphine (10 mg/kg) or with the kappa-opioid agonist U50,488H (U50, 12.5 mg/kg) with or without the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 (0.1 mg/kg). On the day of birth, XX mice showed faster baseline latencies than XY in tail withdrawal, irrespective of their gonadal type. Gonadal males showed greater effects of morphine than gonadal females in the hotplate test, irrespective of their sex chromosome complement. U50 and morphine were effective analgesics in both tests, but MK-801 did not block the U50 effect. The results suggest that sex chromosome complement and gonadal secretions both contribute to sex differences in nociception and analgesia by the day of birth. PERSPECTIVE: Sex differences in pain may stem not only from the action of gonadal hormones on pain circuits but from the sex-specific action of X and Y genes. Identification of sex chromosome genes causing sex differences could contribute to better pain therapy in females and males.


Assuntos
Analgésicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Dor/prevenção & controle , Cromossomos Sexuais/genética , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/administração & dosagem , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgesia/psicologia , Analgésicos/administração & dosagem , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Maleato de Dizocilpina/administração & dosagem , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Knockout , Morfina/administração & dosagem , Morfina/farmacologia , Dor/fisiopatologia , Dor/psicologia , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Caracteres Sexuais , Processos de Determinação Sexual , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA