Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neurosci Res ; 100(1): 19-34, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32830380

RESUMO

The actions of endogenous opioids and nociceptin/orphanin FQ are mediated by four homologous G protein-coupled receptors that constitute the opioid receptor family. However, little is known about opioid systems in cyclostomes (living jawless fish) and how opioid systems might have evolved from invertebrates. Here, we leveraged de novo transcriptome and low-coverage whole-genome assembly in the Pacific hagfish (Eptatretus stoutii) to identify and characterize the first full-length coding sequence for a functional opioid receptor in a cyclostome. Additionally, we define two novel endogenous opioid precursors in this species that predict several novel opioid peptides. Bioinformatic analysis shows no closely related opioid receptor genes in invertebrates with regard either to the genomic organization or to conserved opioid receptor-specific sequences that are common in all vertebrates. Furthermore, no proteins analogous to vertebrate opioid precursors could be identified by genomic searches despite previous claims of protein or RNA-derived sequences in several invertebrate species. The presence of an expressed orthologous receptor and opioid precursors in the Pacific hagfish confirms that a functional opioid system was likely present in the common ancestor of all extant vertebrates some 550 million years ago, earlier than all previous authenticated accounts. We discuss the premise that the cyclostome and vertebrate opioid systems evolved from invertebrate systems concerned with antimicrobial defense and speculate that the high concentrations of opioid precursors in tissues such as the testes, gut, and activated immune cells are key remnants of this evolutionary role.


Assuntos
Feiticeiras (Peixe) , Analgésicos Opioides , Animais , Evolução Biológica , Evolução Molecular , Feiticeiras (Peixe)/genética , Peptídeos Opioides , Filogenia
2.
J Neurosci ; 35(22): 8442-50, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041913

RESUMO

Chronic pain attenuates midbrain dopamine (DA) transmission, as evidenced by a decrease in opioid-evoked DA release in the ventral striatum, suggesting that the occurrence of chronic pain impairs reward-related behaviors. However, mechanisms by which pain modifies DA transmission remain elusive. Using in vivo microdialysis and microinjection of drugs into the mesolimbic DA system, we demonstrate in mice and rats that microglial activation in the VTA compromises not only opioid-evoked release of DA, but also other DA-stimulating drugs, such as cocaine. Our data show that loss of stimulated extracellular DA is due to impaired chloride homeostasis in midbrain GABAergic interneurons. Treatment with minocycline or interfering with BDNF signaling restored chloride transport within these neurons and recovered DA-dependent reward behavior. Our findings demonstrate that a peripheral nerve injury causes activated microglia within reward circuitry that result in disruption of dopaminergic signaling and reward behavior. These results have broad implications that are not restricted to the problem of pain, but are also relevant to affective disorders associated with disruption of reward circuitry. Because chronic pain causes glial activation in areas of the CNS important for mood and affect, our findings may translate to other disorders, including anxiety and depression, that demonstrate high comorbidity with chronic pain.


Assuntos
Dor Crônica/patologia , Sistema Límbico/patologia , Microglia/patologia , Rede Nervosa/patologia , Recompensa , Animais , Área Sob a Curva , Dor Crônica/tratamento farmacológico , Dor Crônica/etiologia , Cocaína/uso terapêutico , Condicionamento Clássico/efeitos dos fármacos , Modelos Animais de Doenças , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Minociclina/uso terapêutico , Morfina/uso terapêutico , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/complicações , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
3.
Dev Neurobiol ; 76(9): 1003-13, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26663679

RESUMO

Dendritic morphology is a critical determinant of neuronal connectivity, and in postganglionic sympathetic neurons, tonic activity correlates directly with the size of the dendritic arbor. Thus, identifying signaling mechanisms that regulate dendritic arborization of sympathetic neurons is important to understanding how functional neural circuitry is established and maintained in the sympathetic nervous system. Bone morphogenetic proteins (BMPs) promote dendritic growth in sympathetic neurons; however, downstream signaling events that link BMP receptor activation to dendritic growth are poorly characterized. We previously reported that BMP7 upregulates p75(NTR) mRNA in cultured sympathetic neurons. This receptor is implicated in controlling dendritic growth in central neurons but whether p75(NTR) regulates dendritic growth in peripheral neurons is not known. Here, we demonstrate that BMP7 increases p75(NTR) protein in cultured sympathetic neurons, and this effect is blocked by pharmacologic inhibition of signaling via BMP type I receptor. BMP7 does not trigger dendritic growth in sympathetic neurons dissociated from superior cervical ganglia (SCG) of p75(NTR) nullizygous mice, and overexpression of p75(NTR) in p75(NTR) -/- neurons is sufficient to cause dendritic growth even in the absence of BMP7. Morphometric analyses of SCG from wild-type versus p75(NTR) nullizygous mice at 3, 6, and 12 to 16 weeks of age indicated that genetic deletion of p75(NTR) does not prevent dendritic growth but does stunt dendritic maturation in sympathetic neurons. These data support the hypotheses that p75(NTR) is involved in downstream signaling events that mediate BMP7-induced dendritic growth in sympathetic neurons, and suggest that p75(NTR) signaling positively modulates dendritic complexity in sympathetic neurons in vivo. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1003-1013, 2016.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Dendritos/fisiologia , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/fisiologia , Gânglio Cervical Superior/metabolismo , Animais , Dendritos/metabolismo , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento , Receptores de Fator de Crescimento Neural/genética
4.
Neuropsychopharmacology ; 41(4): 949-59, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26202104

RESUMO

Opioid dependence is accompanied by neuroplastic changes in reward circuitry leading to a negative affective state contributing to addictive behaviors and risk of relapse. The current study presents a neuroimmune mechanism through which chronic opioids disrupt the ventral tegmental area (VTA) dopaminergic circuitry that contributes to impaired reward behavior. Opioid dependence was induced in rodents by treatment with escalating doses of morphine. Microglial activation was observed in the VTA following spontaneous withdrawal from chronic morphine treatment. Opioid-induced microglial activation resulted in an increase in brain-derived neurotrophic factor (BDNF) expression and a reduction in the expression and function of the K(+)Cl(-) co-transporter KCC2 within VTA GABAergic neurons. Inhibition of microglial activation or interfering with BDNF signaling prevented the loss of Cl(-) extrusion capacity and restored the rewarding effects of cocaine in opioid-dependent animals. Consistent with a microglial-derived BDNF-induced disruption of reward, intra-VTA injection of BDNF or a KCC2 inhibitor resulted in a loss of cocaine-induced place preference in opioid-naïve animals. The loss of the extracellular Cl(-) gradient undermines GABAA-mediated inhibition, and represents a mechanism by which chronic opioid treatments can result in blunted reward circuitry. This study directly implicates microglial-derived BDNF as a negative regulator of reward in opioid-dependent states, identifying new therapeutic targets for opiate addictive behaviors.


Assuntos
Cocaína/administração & dosagem , Neurônios GABAérgicos/metabolismo , Microglia/metabolismo , Morfina/administração & dosagem , Síndrome de Abstinência a Substâncias/imunologia , Núcleos Ventrais do Tálamo/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios GABAérgicos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Modelos Neurológicos , Neuroimunomodulação , Recompensa , Simportadores/metabolismo , Núcleos Ventrais do Tálamo/efeitos dos fármacos , Cotransportadores de K e Cl-
5.
Toxicol Sci ; 138(2): 379-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24385416

RESUMO

We recently demonstrated that polychlorinated biphenyl (PCB) congeners with multiple ortho chlorine substitutions sensitize ryanodine receptors (RyRs), and this activity promotes Ca²âº-dependent dendritic growth in cultured neurons. Many ortho-substituted congeners display axial chirality, and we previously reported that the chiral congener PCB 136 (2,2',3,3',6,6'-hexachlorobiphenyl) atropselectively sensitizes RyRs. Here, we test the hypothesis that PCB 136 atropisomers differentially alter dendritic growth and other parameters of neuronal connectivity influenced by RyR activity. (-)-PCB 136, which potently sensitizes RyRs, enhances dendritic growth in primary cultures of rat hippocampal neurons, whereas (+)-PCB 136, which lacks RyR activity, has no effect on dendritic growth. The dendrite-promoting activity of (-)-PCB 136 is observed at concentrations ranging from 0.1 to 100 nM and is blocked by pharmacologic RyR antagonism. Neither atropisomer alters axonal growth or cell viability. Quantification of PCB 136 atropisomers in hippocampal cultures indicates that atropselective effects on dendritic growth are not due to differential partitioning of atropisomers into cultured cells. Imaging of hippocampal neurons loaded with Ca²âº-sensitive dye demonstrates that (-)-PCB 136 but not (+)-PCB 136 increases the frequency of spontaneous Ca²âº oscillations. Similarly, (-)-PCB 136 but not (+)-PCB 136 increases the activity of hippocampal neurons plated on microelectrode arrays. These data support the hypothesis that atropselective effects on RyR activity translate into atropselective effects of PCB 136 atropisomers on neuronal connectivity, and suggest that the variable atropisomeric enrichment of chiral PCBs observed in the human population may be a significant determinant of individual susceptibility for adverse neurodevelopmental outcomes following PCB exposure.


Assuntos
Poluentes Ambientais/toxicidade , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Bifenilos Policlorados/toxicidade , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Sinalização do Cálcio/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Cones de Crescimento/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Microeletrodos , Neurônios/metabolismo , Neurônios/patologia , Bifenilos Policlorados/química , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Estereoisomerismo
6.
J Vis Exp ; (61)2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22473299

RESUMO

The shape of the dendritic arbor determines the total synaptic input a neuron can receive (1-3), and influences the types and distribution of these inputs (4-6). Altered patterns of dendritic growth and plasticity are associated with impaired neurobehavioral function in experimental models (7), and are thought to contribute to clinical symptoms observed in both neurodevelopmental disorders (8-10) and neurodegenerative diseases (11-13). Such observations underscore the functional importance of precisely regulating dendritic morphology, and suggest that identifying mechanisms that control dendritic growth will not only advance understanding of how neuronal connectivity is regulated during normal development, but may also provide insight on novel therapeutic strategies for diverse neurological diseases. Mechanistic studies of dendritic growth would be greatly facilitated by the availability of a model system that allows neurons to be experimentally switched from a state in which they do not extend dendrites to one in which they elaborate a dendritic arbor comparable to that of their in vivo counterparts. Primary cultures of sympathetic neurons dissociated from the superior cervical ganglia (SCG) of perinatal rodents provide such a model. When cultured in defined medium in the absence of serum and ganglionic glial cells, sympathetic neurons extend a single process which is axonal, and this unipolar state persists for weeks to months in culture (14,15). However, the addition of either bone morphogenetic protein-7 (BMP-7) (16,17) or Matrigel (18) to the culture medium triggers these neurons to extend multiple processes that meet the morphologic, biochemical and functional criteria for dendrites. Sympathetic neurons dissociated from the SCG of perinatal rodents and grown under defined conditions are a homogenous population of neurons (19) that respond uniformly to the dendrite-promoting activity of Matrigel, BMP-7 and other BMPs of the decapentaplegic (dpp) and 60A subfamilies (17,18,20,21). Importantly, Matrigel- and BMP-induced dendrite formation occurs in the absence of changes in cell survival or axonal growth (17,18). Here, we describe how to set up dissociated cultures of sympathetic neurons derived from the SCG of perinatal rats so that they are responsive to the selective dendrite-promoting activity of Matrigel or BMPs.


Assuntos
Técnicas de Cultura de Células/métodos , Dendritos/fisiologia , Neurônios/citologia , Gânglio Cervical Superior/citologia , Animais , Proteína Morfogenética Óssea 7 , Processos de Crescimento Celular/fisiologia , Colágeno , Meios de Cultura , Combinação de Medicamentos , Laminina , Proteoglicanas , Ratos
7.
Mol Biol Cell ; 22(19): 3734-46, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21849478

RESUMO

The receptor Deleted in Colorectal Cancer (DCC) mediates the attractive response of axons to the guidance cue netrin-1 during development. On netrin-1 stimulation, DCC is phosphorylated and induces the assembly of signaling complexes within the growth cone, leading to activation of cytoskeleton regulators, namely the GTPases Rac1 and Cdc42. The molecular mechanisms that link netrin-1/DCC to the actin machinery remain unclear. In this study we seek to demonstrate that the actin-binding proteins ezrin-radixin-moesin (ERM) are effectors of netrin-1/DCC signaling in embryonic cortical neurons. We show that ezrin associates with DCC in a netrin-1-dependent manner. We demonstrate that netrin-1/DCC induces ERM phosphorylation and activation and that the phosphorylation of DCC is required in that context. Moreover, Src kinases and RhoA/Rho kinase activities mediate netrin-1-induced ERM phosphorylation in neurons. We also observed that phosphorylated ERM proteins accumulate in growth cone filopodia, where they colocalize with DCC upon netrin-1 stimulation. Finally, we show that loss of ezrin expression in cortical neurons significantly decreases axon outgrowth induced by netrin-1. Together, our findings demonstrate that netrin-1 induces the formation of an activated ERM/DCC complex in growth cone filopodia, which is required for netrin-1-dependent cortical axon outgrowth.


Assuntos
Axônios/metabolismo , Sistema Nervoso Central/embriologia , Proteínas do Citoesqueleto/metabolismo , Desenvolvimento Embrionário/genética , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Quinases da Família src/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Proteínas do Citoesqueleto/genética , Citoesqueleto/metabolismo , Receptor DCC , Cones de Crescimento/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Complexos Multiproteicos , Fatores de Crescimento Neural/genética , Netrina-1 , Neurônios/citologia , Pseudópodes/genética , Pseudópodes/metabolismo , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/genética
8.
Environ Health Perspect ; 119(4): 519-26, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21106467

RESUMO

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are widely used flame retardants that bioaccumulate in human tissues. Their neurotoxicity involves dysregulation of calcium ion (Ca(2+))signaling; however, specific mechanisms have yet to be defined. OBJECTIVE: We aimed to define the structure-activity relationship (SAR) for PBDEs and their metabolites toward ryanodine receptors type 1 (RyR1) and type 2 (RyR2) and to determine whether it predicts neurotoxicity. METHODS: We analyzed [3H]ryanodine binding, microsomal Ca(2+) fluxes, cellular measurements of Ca(2+) homeostasis, and neurotoxicity to define mechanisms and specificity of PBDE-mediated Ca(2+) dysregulation. RESULTS: PBDEs possessing two ortho-bromine substituents and lacking at least one para-bromine substituent (e.g., BDE-49) activate RyR1 and RyR2 with greater efficacy than corresponding congeners with two para-bromine substitutions (e.g., BDE-47). Addition of a methoxy group in the free para position reduces the activity of parent PBDEs. The hydroxylated BDEs 6-OH-BDE-47 and 4´-OH-BDE-49 are biphasic RyR modulators. Pretreatment of HEK293 cells (derived from human embryonic kidney cells) expressing either RyR1 or RyR2 with BDE-49 (250 nM) sensitized Ca2+ flux triggered by RyR agonists, whereas BDE-47 (250 nM) had negligible activity. The divergent activity of BDE-49, BDE-47, and 6-OH-BDE-47 toward RyRs predicted neurotoxicity in cultures of cortical neurons. CONCLUSIONS: We found that PBDEs are potent modulators of RyR1 and RyR2. A stringent SAR at the ortho and para position determined whether a congener enhanced, inhibited, or exerted nonmonotonic actions toward RyRs. These results identify a convergent molecular target of PBDEs previously identified for noncoplanar polychlorinated biphenyls (PCBs) that predicts their cellular neurotoxicity and therefore could be a useful tool in risk assessment of PBDEs and related compounds.


Assuntos
Poluentes Ambientais/toxicidade , Éteres Difenil Halogenados/toxicidade , Sistema Nervoso/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Poluentes Ambientais/química , Poluentes Ambientais/metabolismo , Éteres Difenil Halogenados/química , Éteres Difenil Halogenados/metabolismo , Humanos , Sistema Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
9.
Curr Neurobiol ; 2(1): 23-29, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23226931

RESUMO

M2 muscarinic receptors are expressed on both parasympathetic and sympathetic nerve endings where they function as autoinhibitory receptors to limit release of acetylcholine and norepinephrine, respectively. M2 muscarinic receptor expression on parasympathetic nerves is decreased by viral infection and by gamma-interferon (IFNγ) and increased by dexamethasone; and these effects are of clinical relevance in the etiology and treatment of asthma. Whether IFNγ and dexamethasone similarly modulate M2 receptor expression on sympathetic nerves is not known. To address this question, we examined the effects of IFNγ and dexamethasone on M2 receptor expression at the mRNA and protein level in primary cultures of sympathetic neurons dissociated from the rat superior cervical ganglia (SCG). Semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) indicated that neither IFNγ nor dexamethasone altered M2 receptor transcript levels. However, western blot analyses demonstrated that IFNγ, but not dexamethasone, increases M2 receptor protein expression in sympathetic neurons. Increased expression did not significantly alter subcellular localization of M2 receptors in sympathetic neurons as determined using immunocytochemistry. These findings indicate that M2 receptors are differentially regulated in different types of autonomic neurons, and they suggest a novel mechanism by which IFNγ may contribute to airway hyperreactivity in viral-induced asthma.

10.
Mol Cell Biol ; 28(7): 2314-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18212043

RESUMO

The chemotropic guidance cue netrin-1 promotes neurite outgrowth through its receptor Deleted in Colorectal Cancer (DCC) via activation of Rac1. The guanine nucleotide exchange factor (GEF) linking netrin-1/DCC to Rac1 activation has not yet been identified. Here, we show that the RhoGEF Trio mediates Rac1 activation in netrin-1 signaling. We found that Trio interacts with the netrin-1 receptor DCC in mouse embryonic brains and that netrin-1-induced Rac1 activation in brain is impaired in the absence of Trio. Trio(-/-) cortical neurons fail to extend neurites in response to netrin-1, while they are able to respond to glutamate. Accordingly, netrin-1-induced commissural axon outgrowth is reduced in Trio(-/-) spinal cord explants, and the guidance of commissural axons toward the floor plate is affected by the absence of Trio. The anterior commissure is absent in Trio-null embryos, and netrin-1/DCC-dependent axonal projections that form the internal capsule and the corpus callosum are defective in the mutants. Taken together, these findings establish Trio as a GEF that mediates netrin-1 signaling in axon outgrowth and guidance through its ability to activate Rac1.


Assuntos
Cones de Crescimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fatores de Crescimento Neural/fisiologia , Neuropeptídeos/fisiologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Encéfalo/anormalidades , Encéfalo/citologia , Encéfalo/embriologia , Células COS , Linhagem Celular , Células Cultivadas/citologia , Chlorocebus aethiops , Receptor DCC , Ativação Enzimática , Feminino , Cones de Crescimento/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Netrina-1 , Proteínas Oncogênicas/fisiologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Medula Espinal/citologia , Medula Espinal/embriologia , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA