Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Brain Behav Immun ; 62: 53-63, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28179107

RESUMO

Activation of the hypothalamo-pituitary-adrenal (HPA) axis by inflammatory stressors (e.g., bacterial lipopolysaccharide) is thought to involve vascular transduction of circulating cytokines, with perivascular macrophages (PVMs) along with endothelia, effecting activation of HPA control circuitry via inducible (cyclooxygenase-2- or COX-2-dependent) prostaglandin synthesis. To test the stressor-specificity of this mechanism, we examined whether ablation of PVMs or pharmacologic blockade of COX activity affected HPA responses to a representative emotional stressor, restraint. Exposing rats to a single 30min acute restraint episode provoked increased plasma levels of at least one proinflammatory cytokine, IL-6, microglial activation and multiple indices of cerebrovascular activation, including COX-2 expression and increased brain prostaglandin E2 levels at 0-2h after stress. Pretreatment with the nonselective COX inhibitor, indomethacin, either icv (10µg in 5µl) or iv (1mg/kg) significantly reduced restraint-induced Fos expression in the paraventricular hypothalamic nucleus (PVH) by 45%, relative to vehicle-injected controls. A 75% reduction of the PVH activational response was seen in rats exposed to acute restraint 5-7days after ablation of brain PVMs by icv injection of liposomes encapsulating the bisphosphonate drug, clodronate. Basal plasma levels of ACTH and corticosterone were not altered in clodronate liposome-injected rats, but the peak magnitude of restraint-induced HPA secretory responses was substantially reduced, relative to animals pretreated with saline-filled liposomes. These findings support an unexpectedly prominent role for inducible prostaglandin synthesis by PVMs in HPA responses to acute restraint, a prototypic emotional stressor.


Assuntos
Encéfalo/metabolismo , Inflamação/metabolismo , Estresse Fisiológico/fisiologia , Estresse Psicológico/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Inibidores de Ciclo-Oxigenase/farmacologia , Emoções/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Indometacina/farmacologia , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Ratos , Ratos Sprague-Dawley , Restrição Física , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estresse Psicológico/fisiopatologia
2.
J Neuroinflammation ; 13(1): 178, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27391474

RESUMO

BACKGROUND: Neurodegeneration is believed to be the primary cause of permanent, long-term disability in patients with multiple sclerosis. The cause of neurodegeneration in multiple sclerosis appears to be multifactorial. One mechanism that has been implicated in the pathogenesis of neurodegeneration in multiple sclerosis is the targeting of neuronal and axonal antigens by autoantibodies. Multiple sclerosis patients develop antibodies to the RNA-binding protein, heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), which is enriched in neurons. We hypothesized that anti-hnRNP A1 antibodies would contribute to neurodegeneration in an animal model of multiple sclerosis. METHODS: Following induction of experimental autoimmune encephalomyelitis (EAE) by direct immunization with myelin oligodendrocyte glycoprotein, mice were injected with anti-hnRNP A1 or control antibodies. Animals were examined clinically, and the central nervous system (CNS) tissues were tested for neurodegeneration with Fluoro-Jade C, a marker of degenerating neural elements. RESULTS: Injection of anti-hnRNP A1 antibodies in mice with EAE worsened clinical disease, altered the clinical disease phenotype, and caused neurodegeneration preferentially in the ventral spinocerebellar tract and deep white matter of the cerebellum in the CNS. Neurodegeneration in mice injected with hnRNP A1-M9 antibodies compared to control groups was consistent with "dying back" axonal degeneration. CONCLUSIONS: These data suggest that antibodies to the RNA-binding protein hnRNP A1 contribute to neurodegeneration in immune-mediated disease of the CNS.


Assuntos
Autoanticorpos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/imunologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Animais , Autoanticorpos/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/metabolismo
3.
Alzheimers Dement ; 12(5): 527-37, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26555315

RESUMO

INTRODUCTION: Stress and corticotropin-releasing factor (CRF) have been implicated as mechanistically involved in Alzheimer's disease (AD), but agents that impact CRF signaling have not been carefully tested for therapeutic efficacy or long-term safety in animal models. METHODS: To test whether antagonism of the type-1 corticotropin-releasing factor receptor (CRFR1) could be used as a disease-modifying treatment for AD, we used a preclinical prevention paradigm and treated 30-day-old AD transgenic mice with the small-molecule, CRFR1-selective antagonist, R121919, for 5 months, and examined AD pathologic and behavioral end points. RESULTS: R121919 significantly prevented the onset of cognitive impairment in female mice and reduced cellular and synaptic deficits and beta amyloid and C-terminal fragment-ß levels in both genders. We observed no tolerability or toxicity issues in mice treated with R121919. DISCUSSION: CRFR1 antagonism presents a viable disease-modifying therapy for AD, recommending its advancement to early-phase human safety trials.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Cognição/fisiologia , Modelos Animais de Doenças , Receptores de Hormônio Liberador da Corticotropina , Sinapses/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Camundongos , Camundongos Transgênicos , Pirimidinas , Receptores de Hormônio Liberador da Corticotropina/deficiência , Receptores de Hormônio Liberador da Corticotropina/genética
4.
J Pharmacol Exp Ther ; 353(2): 307-17, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25736419

RESUMO

The corticotropin-releasing factor (CRF) peptide family comprises the mammalian peptides CRF and the urocortins as well as frog skin sauvagine and fish urophyseal urotensin. Advances in understanding the roles of the CRF ligand family and associated receptors have often relied on radioreceptor assays using labeled CRF ligands. These assays depend on stable, high-affinity CRF analogs that can be labeled, purified, and chemically characterized. Analogs of several of the native peptides have been used in this context, most prominently including sauvagine from the frog Phyllomedusa sauvageii (PS-Svg). Because each of these affords both advantages and disadvantages, new analogs with superior properties would be welcome. We find that a sauvagine-like peptide recently isolated from a different frog species, Pachymedusa dacnicolor (PD-Svg), is a high-affinity agonist whose radioiodinated analog, [(125)ITyr(0)-Glu(1), Nle(17)]-PD-Svg, exhibits improved biochemical properties over those of earlier iodinated agonists. Specifically, the PD-Svg radioligand binds both CRF receptors with comparably high affinity as its PS-Svg counterpart, but detects a greater number of sites on both type 1 and type 2 receptors. PD-Svg is also ∼10 times more potent at stimulating cAMP accumulation in cells expressing the native receptors. Autoradiographic localization using the PD-Svg radioligand shows robust specific binding to rodent brain and peripheral tissues that identifies consensus CRF receptor-expressing sites in a greater number and/or with greater sensitivity than its PS-Svg counterpart. We suggest that labeled analogs of PD-Svg may be useful tools for biochemical, structural, pharmacological, and anatomic studies of CRF receptors.


Assuntos
Proteínas de Anfíbios/metabolismo , Anuros , Hormônios Peptídicos/metabolismo , Ensaio Radioligante/métodos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Sequência de Aminoácidos , Proteínas de Anfíbios/química , Animais , Linhagem Celular , Humanos , Marcação por Isótopo , Cinética , Ligantes , Camundongos , Dados de Sequência Molecular , Hormônios Peptídicos/química , Transporte Proteico , Ratos , Receptores de Hormônio Liberador da Corticotropina/química
5.
Proc Natl Acad Sci U S A ; 109(16): 6277-82, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22451915

RESUMO

Exposure and/or sensitivity to stress have been implicated as conferring risk for development of Alzheimer's disease (AD). Although the basis for such a link remains unclear, we previously reported differential involvement of corticotropin-releasing factor receptor (CRFR) 1 and 2 in acute stress-induced tau phosphorylation (tau-P) and solubility in the hippocampus. Here we examined the role of CRFRs in tau-P induced by repeated stress and the structural manifestations of altered tau solubility. Robust tau-P responses were seen in WT and CRFR2 null mice exposed to repeated stress, which were sustained at even 24 h after the final stress exposure. A portion of phosphorylated tau in these mice was sequestered in detergent-soluble cellular fractions. In contrast, CRFR1 and CRFR double-KO mice did not exhibit repeated stress-induced alterations in tau-P or solubility. Similarly, treatment with CRFR1 antagonist attenuated repeated stress-induced tau-P. Using histochemical approaches in a transgenic CRFR1 reporter mouse line, we found substantial overlap between hippocampal CRFR1 expression and cells positive for phosphorylated tau after exposure to repeated stress. Ultrastructural analysis of negatively stained extracts from WT and CRFR2 null mice identified globular aggregates that displayed positive immunogold labeling for tau-P, as well as conformational changes in tau (MC1) seen in early AD. Given that repeated stress exposure results in chronic increases in hippocampal tau-P and its sequestration in an insoluble (and potentially prepathogenic) form, our data may define a link between stress and an AD-related pathogenic mechanism.


Assuntos
Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico , Proteínas tau/metabolismo , Animais , Western Blotting , Giro Denteado , Detergentes/química , Feminino , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Imunoeletrônica , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/genética , Solubilidade , Proteínas tau/química , Proteínas tau/ultraestrutura
6.
J Neurosci ; 31(26): 9683-95, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21715634

RESUMO

A network of interconnected limbic forebrain cell groups, including the medial prefrontal cortex (mPFC) and hippocampal formation (HF), is known to shape adaptive responses to emotionally stressful experiences, including output of the hypothalamo-pituitary-adrenal (HPA) axis. While disruption of limbic HPA-inhibitory systems is implicated in stress-related psychiatric and systemic illnesses, progress in the field has been hampered by a lack of a systems-level understanding of the organization that provides for this regulation. Using rats, we first localized cell groups afferent to the paraventricular hypothalamic nucleus (PVH) (the initiator of HPA responses to stress) whose engagement following acute (30 min) restraint was diminished by excitotoxin lesions of the ventral subiculum, a component of the HF. This identified a candidate relay for imparting HF influences in a circumscribed portion of the anterior bed nucleus of the stria terminalis (aBST), which we previously identified as a GABAergic relay subserving mPFC inhibition of the stress axis. Anatomical tracing experiments then indicated that extrinsic projections from HF and mPFC converge onto regions of aBST that contain neurons that are both stress sensitive and PVH projecting. Two final experiments provided evidence that (1) HPA-inhibitory influences of mPFC and HF are additive and (2) aBST plays a more prominent inhibitory role than ventral subiculum over stress-induced HPA endpoints. These findings support the view that stress-inhibitory influences of mPFC and HF are exerted principally via convergence onto a common relay, as opposed to a serial, parallel, or more complex multisynaptic network.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Córtex Pré-Frontal/metabolismo , Estresse Fisiológico/fisiologia , Animais , Sistema Hipotálamo-Hipofisário/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/metabolismo
7.
J Neurosci ; 31(48): 17612-21, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-22131422

RESUMO

CCAAT enhancer binding protein-delta (C/EBPδ) is a transcription factor that regulates inflammatory processes mediating bystander neuronal injury and CNS autoimmune inflammatory disease. The mechanism of the involvement of C/EBPδ in these processes remains to be determined. Here, we examined the cellular source(s) and mechanisms by which C/EBPδ may be involved in an animal model of multiple sclerosis. Mice deficient in C/EBPδ expression exhibited less severe clinical disease than wild-type littermates in response to induction of experimental autoimmune encephalomyelitis (EAE) by vaccination with a myelin oligodendrocyte glycoprotein (MOG) fragment. This reduction in EAE severity was associated with a significant alteration in the complement of major CNS T-helper (Th) cell subtypes throughout disease, manifest as reduced ratios of Th17 cells to regulatory T-cells (Tregs). Studies in bone marrow chimeric mice indicated that C/EBPδ expression by peripherally derived immune cells mediates C/EBPδ involvement in EAE. Follow up in vitro and in vivo examination of dendritic cell (DC) mediated Th-cell development suggests that C/EBPδ suppresses DC expression of interleukin-10 (IL-10), favoring Th17 over Treg development. In vitro and in vivo blockade of IL-10 signaling attenuated the effect of reduced C/EBPδ expression by DCs on Th17:Treg ratios. These findings identify C/EBPδ as an important DC transcription factor in CNS autoimmune inflammatory disease by virtue of its capacity to alter the Th17:Treg balance in an IL-10 dependent fashion.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/genética , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Interleucina-10/metabolismo , Camundongos , Camundongos Knockout , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Associada a Mielina/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Regulação para Cima
8.
Int J Mol Sci ; 14(1): 547-62, 2012 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-23271370

RESUMO

Multiple sclerosis (MS) is a devastating neurological disease that predominantly affects young adults resulting in severe personal and economic impact. The majority of therapies for this disease were developed in, or are beneficial in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. While known to target adaptive anti-CNS immune responses, they also target, the innate immune arm. This mini-review focuses on the role of dendritic cells (DCs), the professional antigen presenting cells of the innate immune system. The evidence for a role for DCs in the appropriate regulation of anti-CNS autoimmune responses and their role in MS disease susceptibility and possible therapeutic utility are discussed. Additionally, the current controversy regarding the evidence for the presence of functional DCs in the normal CNS is reviewed. Furthermore, the role of CNS DCs and potential routes of their intercourse between the CNS and cervical lymph nodes are considered. Finally, the future role that this nexus between the CNS and the cervical lymph nodes might play in site directed molecular and cellular therapy for MS is outlined.


Assuntos
Células Dendríticas/imunologia , Tolerância Imunológica , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia , Animais , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Terapia de Alvo Molecular , Vitamina D/metabolismo
9.
Eur J Neurosci ; 34(3): 448-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21722209

RESUMO

Clinical studies suggest that exposure to stress can increase risk for Alzheimer's disease (AD). Although the precise links between stress and vulnerability to develop AD remain uncertain, recent animal work suggests that stress may promote susceptibility to AD pathology by activating tau kinases and inducing tau phosphorylation (tau-P). Our previous findings indicate the differential involvement of corticotropin-releasing factor receptor (CRFR) types 1 and 2 in regulating tau-P in the hippocampus induced by acute restraint, an emotional stressor. To assess the generality of CRFR involvement in stress-induced tau-P and tau kinase activity, the present study extends our investigation to a well-characterized physiological stressor, i.e. immune challenge induced by bacterial lipopolysaccharide (LPS). Acute systemic administration of LPS (100 µg/kg) robustly increased hippocampal (but not isocortical or cerebellar) tau-P, peaking at 40-120 min postinjection and abating thereafter. Assessments of the genotype dependence of this effect yielded results that were distinct from the restraint model. Treatment with LPS increased phosphorylation in wild-type, single and double CRFR knockouts with only subtle variation, which included a reliable exaggeration of tau-P responses in CRFR1-deficient mice. Parallel analyses implicated glycogen synthase kinase-3 and cyclin-dependent kinase-5 as likely cellular mediators of LPS-induced tau-P. Conversely, our data suggest that temperature-dependent fluctuations in tau protein phosphatase 2A (PP2A) may not play a role in this context. Thus, neither the strict CRFR1 dependence of restraint-induced tau-P nor the exaggeration of these responses in CRFR2 null mice generalize to the LPS model. CRFR mediation of stress-induced hippocampal tau-P may be limited to emotional stressors.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Lipopolissacarídeos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Proteínas tau/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Ativação Enzimática , Feminino , Hipocampo/citologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Fisiológico , Estresse Psicológico
10.
Nat Neurosci ; 10(10): 1249-59, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17828261

RESUMO

G protein-gated potassium (Kir3) channels are important for controlling neuronal excitability in the brain. Using a proteomics approach, we have identified a unique rodent intracellular protein, sorting nexin 27 (SNX27), which regulates the trafficking of Kir3 channels. Like most sorting nexins, SNX27 possesses a functional PX domain that selectively binds the membrane phospholipid phosphatidylinositol-3-phosphate (PI3P) and is important for trafficking to the early endosome. SNX27, however, is the only sorting nexin to contain a PDZ domain. This PDZ domain discriminates between channels with similar class I PDZ-binding motifs, associating with the C-terminal end of Kir3.3 and Kir3.2c (-ESKV), but not with that of Kir2.1 (-ESEI) or Kv1.4 (-ETDV). SNX27 promotes the endosomal movement of Kir3 channels, leading to reduced surface expression, increased degradation and smaller Kir3 potassium currents. The regulation of endosomal trafficking via sorting nexins reveals a previously unknown mechanism for controlling potassium channel surface expression.


Assuntos
Encéfalo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Domínios PDZ/fisiologia , Animais , Encéfalo/citologia , Linhagem Celular Transformada , Endocitose/fisiologia , Regulação da Expressão Gênica/genética , Humanos , Imunoprecipitação/métodos , Masculino , Potenciais da Membrana/genética , Dados de Sequência Molecular , Técnicas de Patch-Clamp/métodos , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Proteômica , Ratos , Transfecção/métodos
11.
J Neurosci ; 29(41): 12970-81, 2009 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-19828811

RESUMO

Systemic injection of lipopolysaccharide (LPS) is a widely used model of immune/inflammatory challenge, which can invoke a host of CNS responses, including activation of the hypothalamic-pituitary-adrenal (HPA) axis. Inducible vascular prostaglandin E(2) (PGE(2)) synthesis by endothelial (ECs) and/or perivascular cells (PVCs) (a macrophage-derived vascular cell type) is implicated in the engagement of HPA and other CNS responses, by virtue of their capacity to express cyclooxygenase-2 (COX-2) and microsomal PGE(2) synthase-1. Evidence from genetic and pharmacologic studies also supports a role for the constitutively expressed COX-1 in inflammation-induced activation of the HPA axis, although histochemical evidence to support relevant localization(s) and regulation of COX-1 expression is lacking. The present experiments fill this void in showing that COX-1 immunoreactivity (IR) and mRNA are detectable in identified PVCs and parenchymal microglia under basal conditions and is robustly expressed in these and ECs 1-3 h after intravenous injection of LPS (2 microg/kg). Confocal and electron microscopic analyses indicate distinct cellular/subcellular localizations of COX-1-IR in the three cell types. Interestingly, COX-1 expression is enhanced in ECs of brain PVC-depleted rats, supporting an anti-inflammatory role of the latter cell type. Functional involvement of COX-1 is indicated by the observation that central, but not systemic, pretreatment with the selective COX-1 inhibitor SC-560 attenuated the early phase of LPS-induced increases in adrenocorticotropin and corticosterone secretion. These findings support an involvement of COX-1 in bidirectional interplay between ECs and PVCs in initiating vascular PGE(2) and downstream HPA response to proinflammatory challenges.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Inflamação/patologia , Sistema Hipófise-Suprarrenal/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Corticosterona/metabolismo , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/genética , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipotálamo-Hipofisário/ultraestrutura , Técnicas Imunoenzimáticas/métodos , Inflamação/induzido quimicamente , Injeções Intraventriculares/métodos , Interleucina-1beta/administração & dosagem , Lipopolissacarídeos , Lipossomos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão/métodos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiopatologia , Sistema Hipófise-Suprarrenal/ultraestrutura , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de von Willebrand/metabolismo
12.
J Neurosci ; 29(22): 7330-40, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19494154

RESUMO

Complementing its roles in cognitive and affective information processing, the medial prefrontal cortex (mPFC) is a nodal point of a limbic forebrain circuit that modulates stress-related homeostatic mechanisms, including the hypothalamic-pituitary-adrenal (HPA) axis. mPFC influences on HPA output are predominantly inhibitory and emanate from the prelimbic and/or dorsal anterior cingulate cortical fields (PL and ACd, respectively). mPFC projections do not target HPA effector neurons in the paraventricular hypothalamic nucleus (PVH) directly, distributing instead to nearby forebrain regions, including some that house GABAergic neurons implicated in inhibitory PVH control. To identify pathway(s) subserving HPA-inhibitory mPFC influences, an initial screen for sources of GABAergic input to PVH whose sensitivity to an acute emotional (restraint) stress was diminished by PL/ACd lesions identified a discrete region of the anterior bed nucleus of the stria terminalis (aBST) as a candidate for fulfilling this role. Anatomical tracing experiments confirmed projections from PL (but not ACd) to implicated aBST cell groups, and from these to PVH. Finally, selective immunotoxin-mediated ablation of GABAergic aBST neurons recapitulated the effects of PL/ACd lesions on acute stress-induced activation of HPA output. The identification of a proximate mediator of HPA-inhibitory limbic influences provides a framework for clarifying how inhibitory neural and hormonal controls of HPA output are integrated, adaptations of the axis to chronic stress are effected, and how endocrine abnormalities may contribute to stress-related psychiatric illnesses in which mPFC dysfunction is implicated.


Assuntos
Inibição Neural/fisiologia , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Córtex Pré-Frontal/fisiologia , Estresse Psicológico/fisiopatologia , Ácido gama-Aminobutírico/metabolismo , Análise de Variância , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Dextranos/metabolismo , Agonistas de Aminoácidos Excitatórios/toxicidade , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Ácido Ibotênico/toxicidade , Masculino , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Oncogênicas v-fos/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Fito-Hemaglutininas/farmacologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/lesões , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Estilbamidinas/metabolismo
13.
J Neurosci ; 29(1): 118-30, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19129390

RESUMO

Axonal dysfunction is the major phenotypic change in many neurodegenerative diseases, but the processes underlying this impairment are not clear. Modifier of cell adhesion (MOCA) is a presenilin binding protein that functions as a guanine nucleotide exchange factor for Rac1. The loss of MOCA in mice leads to axonal degeneration and causes sensorimotor impairments by decreasing cofilin phosphorylation and altering its upstream signaling partners LIM kinase and p21-activated kinase, an enzyme directly downstream of Rac1. The dystrophic axons found in MOCA-deficient mice are associated with abnormal aggregates of neurofilament protein, the disorganization of the axonal cytoskeleton, and the accumulation of autophagic vacuoles and polyubiquitinated proteins. Furthermore, MOCA deficiency causes an alteration in the actin cytoskeleton and the formation of cofilin-containing rod-like structures. The dystrophic axons show functional abnormalities, including impaired axonal transport. These findings demonstrate that MOCA is required for maintaining the functional integrity of axons and define a model for the steps leading to axonal degeneration.


Assuntos
Axônios/patologia , Degeneração Neural/genética , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/deficiência , Transdução de Sinais/genética , Fatores de Despolimerização de Actina/metabolismo , Análise de Variância , Animais , Transporte Axonal/fisiologia , Axônios/metabolismo , Axônios/ultraestrutura , Proteínas de Transporte , Células Cultivadas , Córtex Cerebral/citologia , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Transtornos Neurológicos da Marcha/genética , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão/métodos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Neuropatia Ciática , Medula Espinal/metabolismo , Estilbamidinas/metabolismo , Ubiquitina C/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-32849267

RESUMO

Lactation is a complex physiological process, depending on orchestrated central and peripheral events, including substantial brain plasticity. Among these events is a novel expression of pro-melanin-concentrating hormone (Pmch) mRNA in the rodent hypothalamus, such as the ventral part of the medial preoptic area (vmMPOA). This expression reaches its highest levels around postpartum day 19 (PPD19), when dams transition from lactation to the weaning period. The appearance of this lactation-related Pmch expression occurs simultaneously with the presence of one of the Pmch products, melanin-concentrating hormone (MCH), in the serum. Given the relevance of the MPOA to maternal physiology and the contemporaneity between Pmch expression in this structure and the weaning period, we hypothesized that MCH has a role in the termination of lactation, acting as a mediator between central and peripheral changes. To test this, we investigated the presence of the MCH receptor 1 (MCHR1) and its gene expression in the mammary gland of female rats in different stages of the reproductive cycle. To that end, in situ hybridization, RT-PCR, RT-qPCR, nucleotide sequencing, immunohistochemistry, and Western blotting were employed. Although Mchr1 expression was detected in the epidermis and dermis of both diestrus and lactating rats, parenchymal expression was exclusively found in the functional mammary gland of lactating rats. The expression of Mchr1 mRNA oscillated through the lactation period and reached its maximum in PPD19 dams. Presence of MCHR1 was confirmed with immunohistochemistry with preferential location of MCHR1 immunoreactive cells in the alveolar secretory cells. As was the case for gene expression, the MCHR1 protein levels were significantly higher in PPD19 than in other groups. Our data demonstrate the presence of an anatomical basis for the participation of MCH peptidergic system on the control of lactation through the mammary gland, suggesting that MCH could modulate a prolactation action in early postpartum days and the opposite role at the end of the lactation.


Assuntos
Lactação , Glândulas Mamárias Animais/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Animais , Feminino , Imuno-Histoquímica , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Ratos , Ratos Long-Evans
15.
J Neurosci ; 28(22): 5806-16, 2008 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-18509042

RESUMO

The medial prefrontal cortex (mPFC) has been proposed to play a role in the inhibition of hypothalamo-pituitary-adrenal (HPA) responses to emotional stress via influences on neuroendocrine effector mechanisms housed in the paraventricular hypothalamic nucleus (PVH). Previous work also suggests an involvement of the locus ceruleus (LC) in behavioral and neuroendocrine responses to a variety of acute stressors. The LC issues a widespread set of noradrenergic projections, and its innervation of the prefrontal cortex plays an important role in the modulation of working memory and attention. Because these operations are likely to be critical for stimulus selection, evaluation, and comparison with past experience in mounting adaptive responses to emotional stress, it follows that the LC-to-mPFC pathway might also be involved in regulating HPA activity under such conditions. Therefore, in the present study, we assessed the effects of selectively ablating noradrenergic inputs into the mPFC, using the axonally transported catecholamine immunotoxin, saporin-conjugated antiserum to dopamine-beta-hydroxylase, on acute restraint stress-induced activation of HPA output. Immunotoxin injections in the dorsal mPFC (centered in the prelimbic cortex) attenuated increments in restraint-induced Fos and corticotropin-releasing factor mRNA expression in the neurosecretory region of PVH, as well as HPA secretory responses. Stress-induced Fos expression in dorsal mPFC was enhanced after noradrenergic deafferentation and was negatively correlated with stress-induced PVH activation, independent of lesion status. These findings identify the LC as an upstream component of a circuitry providing for dorsal mPFC modulation of emotional stress-induced HPA activation.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Norepinefrina/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/patologia , Hormônio Adrenocorticotrópico/genética , Hormônio Adrenocorticotrópico/metabolismo , Animais , Química Encefálica/efeitos dos fármacos , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Denervação/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Imunotoxinas/efeitos adversos , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Proteínas Oncogênicas v-fos/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/sangue , Estresse Psicológico/induzido quimicamente
16.
Brain Behav Immun ; 23(7): 1038-52, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19524662

RESUMO

Bacterial lipopolysaccharide (LPS) is widely used to study immune influences on the CNS, and cerebrovascular prostaglandin (PG) synthesis is implicated in mediating LPS influences on some acute phase responses. Other bacterial products, such as staphylococcal enterotoxin B (SEB), impact target tissues differently in that their effects are T-lymphocyte-dependent, yet both LPS and SEB recruit a partially overlapping set of subcortical central autonomic cell groups. We sought to compare neurovascular responses to the two pathogens, and the mechanisms by which they may access the brain. Rats received iv injections of LPS (2 microg/kg), SEB (1mg/kg) or vehicle and were sacrificed 0.5-3h later. Both challenges engaged vascular cells as early 0.5h, as evidenced by induced expression of the vascular early response gene (Verge), and the immediate-early gene, NGFI-B. Cyclooxygenase-2 (COX-2) expression was detected in both endothelial and perivascular cells (PVCs) in response to LPS, but only in PVCs of SEB-challenged animals. The non-selective COX inhibitor, indomethacin (1mg/kg, iv), blocked LPS-induced activation in a subset of central autonomic structures, but failed to alter SEB-driven responses. Liposome mediated ablation of PVCs modulated the CNS response to LPS, did not affect the SEB-induced activational profile. By contrast, disruptions of interoceptive signaling by area postrema lesions or vagotomy (complete or hepatic) markedly attenuated SEB-, but not LPS-, stimulated central activational responses. Despite partial overlap in their neuronal and vascular response profiles, LPS and SEB appear to use distinct mechanisms to access the brain.


Assuntos
Vasos Sanguíneos/imunologia , Encéfalo/imunologia , Ácido Clodrônico/farmacologia , Células Endoteliais/imunologia , Neurônios/imunologia , Linfócitos T/imunologia , Animais , Área Postrema/lesões , Área Postrema/fisiopatologia , Vasos Sanguíneos/metabolismo , Encéfalo/metabolismo , Ciclo-Oxigenase 2/imunologia , Ciclo-Oxigenase 2/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Enterotoxinas/toxicidade , Proteínas Imediatamente Precoces/imunologia , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Indometacina/farmacologia , Injeções Intravenosas , Injeções Intraventriculares , Lipopolissacarídeos/toxicidade , Lipossomos , Ativação Linfocitária/imunologia , Masculino , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/imunologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Linfócitos T/metabolismo , Vagotomia
17.
PLoS Biol ; 4(9): e284, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16933973

RESUMO

All phases of lipopolysaccharide (LPS)-induced fever are mediated by prostaglandin (PG) E2. It is known that the second febrile phase (which starts at approximately 1.5 h post-LPS) and subsequent phases are mediated by PGE2 that originated in endotheliocytes and perivascular cells of the brain. However, the location and phenotypes of the cells that produce PGE2 triggering the first febrile phase (which starts at approximately 0.5 h) remain unknown. By studying PGE2 synthesis at the enzymatic level, we found that it was activated in the lung and liver, but not in the brain, at the onset of the first phase of LPS fever in rats. This activation involved phosphorylation of cytosolic phospholipase A2 (cPLA2) and transcriptional up-regulation of cyclooxygenase (COX)-2. The number of cells displaying COX-2 immunoreactivity surged in the lung and liver (but not in the brain) at the onset of fever, and the majority of these cells were identified as macrophages. When PGE2 synthesis in the periphery was activated, the concentration of PGE2 increased both in the venous blood (which collects PGE2 from tissues) and arterial blood (which delivers PGE2 to the brain). Most importantly, neutralization of circulating PGE2 with an anti-PGE2 antibody both delayed and attenuated LPS fever. It is concluded that fever is initiated by circulating PGE2 synthesized by macrophages of the LPS-processing organs (lung and liver) via phosphorylation of cPLA2 and transcriptional up-regulation of COX-2. Whether PGE2 produced at the level of the blood-brain barrier also contributes to the development of the first phase remains to be clarified.


Assuntos
Febre/induzido quimicamente , Febre/metabolismo , Lipopolissacarídeos/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Febre/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Fígado/citologia , Fígado/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Macrófagos/metabolismo , Masculino , Ratos , Ratos Long-Evans , Transdução de Sinais , Regulação para Cima
18.
J Neurosci ; 27(24): 6552-62, 2007 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-17567816

RESUMO

Hyperphosphorylation of the microtubule-associated protein tau is a key event in the development of Alzheimer's disease (AD) neuropathology. Acute stress can induce hippocampal tau phosphorylation (tau-P) in rodents, but the mechanisms and pathogenic relevance of this response are unclear. Here, we find that hippocampal tau-P elicited by an acute emotional stressor, restraint, was not affected by preventing the stress-induced rise in glucocorticoids but was blocked by genetic or pharmacologic disruption of signaling through the type 1 corticotropin-releasing factor receptor (CRFR1). Conversely, these responses were exaggerated in CRFR2-deficient mice. Parallel CRFR dependence was seen in the stress-induced activation of specific tau kinases. Repeated stress exposure elicited cumulative effects on tau-P and its sequestration in an insoluble, and potentially pathogenic, form. These findings support differential regulatory roles for CRFRs in an AD-relevant form of neuronal plasticity and may link datasets documenting alterations in the CRF signaling system in AD and implicating chronic stress as a risk factor in age-related neurological disorders.


Assuntos
Receptores de Hormônio Liberador da Corticotropina/fisiologia , Estresse Fisiológico/metabolismo , Proteínas tau/metabolismo , Adrenalectomia/métodos , Análise de Variância , Animais , Anticorpos Monoclonais/metabolismo , Corticosterona/administração & dosagem , Hormônio Liberador da Corticotropina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imunoprecipitação/métodos , Injeções Intraventriculares/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Quinases/metabolismo , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/deficiência , Fatores de Tempo
19.
J Neurosci ; 26(50): 12967-76, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17167086

RESUMO

The medial prefrontal cortex (mPFC) is an important neural substrate for integrating cognitive-affective information and regulating the hypothalamo-pituitary-adrenal (HPA) axis response to emotional stress. mPFC modulation of stress responses is effected in part via the paraventricular hypothalamic nucleus (PVH), which houses both autonomic (sympathoadrenal) and neuroendocrine (HPA) effector mechanisms. Although the weight of evidence suggests that mPFC influences on stress-related PVH outputs are inhibitory, discordant findings have been reported, and such work has tended to treat this cortical region as a unitary structure. Here we compared the effects of lesions of the dorsal versus ventral aspects of mPFC, centered in the prelimbic and infralimbic fields, respectively, on acute restraint stress-induced activation of PVH cell groups mediating autonomic and neuroendocrine responses. Lesions to the dorsal mPFC enhanced restraint-induced Fos and corticotropin-releasing factor (CRF) mRNA expression in the neurosecretory region of PVH. Ablation of the ventral mPFC decreased stress-induced Fos protein and CRF mRNA expression in this compartment but increased Fos induction in PVH regions involved in central autonomic control. Repetition of the experiments in rats bearing retrograde tracer deposits to label PVH-autonomic projections confirmed that ventral mPFC lesions selectively increased stress-induced Fos expression in identified preautonomic neurons. Finally, hormonal indices of HPA activation in response to acute stress were augmented after dorsal mPFC lesions and attenuated after ventral mPFC lesions. These results suggest that dorsal and ventral aspects of the mPFC differentially regulate neuroendocrine and autonomic PVH outputs in response to emotional stress.


Assuntos
Adaptação Psicológica/fisiologia , Emoções/fisiologia , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Doença Aguda , Hormônio Adrenocorticotrópico/análise , Hormônio Adrenocorticotrópico/biossíntese , Animais , Hormônio Liberador da Corticotropina/análise , Hormônio Liberador da Corticotropina/biossíntese , Masculino , Rede Nervosa/química , Rede Nervosa/metabolismo , Córtex Pré-Frontal/química , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/psicologia
20.
J Comp Neurol ; 502(2): 236-60, 2007 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-17348011

RESUMO

Murine models of experimental autoimmune encephalomyelitis (EAE) are important vehicles for studying the effects of genetic manipulation on disease processes related to multiple sclerosis (MS). Currently, a comprehensive assessment of EAE pathogenesis with respect to inflammatory and degenerating neuronal elements is lacking. By using Fluoro-jade histochemistry to mark neurodegeneration and dual immunostaining to follow T-cell, microglial, and vascular responses, the time course and distribution of pathological events in EAE was surveyed. C57BL/6J mice were killed at 7, 10, 14, 21 or 35 days after vaccination with the myelin oligodendrocyte glycoprotein peptide MOG(35-55). Disease onset occurred at day 14 and peaked at day 21. Early T-cell infiltration and microglial activation in periventricular and superficial white matter structures adjacent to meninges suggested initial recruitment of effector T cells via the cerebrospinal fluid and choroid plexus. This was associated with microglial activation at distal sites along the same white matter tracts, with subsequent vascular recruitment of T cells associated with further injury. Systematic examination of the entire CNS supported this two-step model of EAE pathogenesis, with inflammation and neurodegeneration commencing at similar times and affecting multiple levels of predominantly sensory central pathways, including their terminal fields. This included aspects of the visual, auditory/vestibular, somatosensory (lemniscal), and proprioceptive (spinocerebellar) systems. The early targeting of visual and periventricular structures followed by more widespread CNS involvement is consistent with common presenting signs in human MS patients and suggestive of a similar basis in neuropathology.


Assuntos
Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/patologia , Inflamação/etiologia , Inflamação/patologia , Degeneração Neural/etiologia , Degeneração Neural/patologia , Animais , Encéfalo/citologia , Encéfalo/ultraestrutura , Encefalomielite Autoimune Experimental/induzido quimicamente , Feminino , Fluoresceínas , Adjuvante de Freund , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão/métodos , Degeneração Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Compostos Orgânicos/metabolismo , Medula Espinal/patologia , Medula Espinal/ultraestrutura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA