Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(12): 113471, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-37980561

RESUMO

Co-transmission of multiple neurotransmitters from a single neuron increases the complexity of signaling information within defined neuronal circuits. Superficial short-axon cells in the olfactory bulb release both dopamine and γ-aminobutyric acid (GABA), yet the specific targets of these neurotransmitters and their respective roles in olfaction have remained unknown. Here, we implement intersectional genetics in mice to selectively block GABA or dopamine release from superficial short-axon cells to identify their distinct cellular targets, impact on circuit function, and behavioral contribution of each neurotransmitter toward olfactory behaviors. We provide functional and anatomical evidence for divergent superficial short-axon cell signaling onto downstream neurons to shape patterns of mitral cell firing that contribute to olfactory-related behaviors.


Assuntos
Bulbo Olfatório , Olfato , Camundongos , Animais , Bulbo Olfatório/fisiologia , Olfato/fisiologia , Dopamina , Interneurônios/fisiologia , Ácido gama-Aminobutírico , Neurotransmissores
2.
Sci Rep ; 12(1): 22044, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36543829

RESUMO

Environmental cues and internal states such as mood, reward, or aversion directly influence feeding behaviors beyond homeostatic necessity. The hypothalamus has been extensively investigated for its role in homeostatic feeding. However, many of the neural circuits that drive more complex, non-homeostatic feeding that integrate valence and sensory cues (such as taste and smell) remain unknown. Here, we describe a basal forebrain (BF)-to-lateral habenula (LHb) circuit that directly modulates non-homeostatic feeding behavior. Using viral-mediated circuit mapping, we identified a population of glutamatergic neurons within the BF that project to the LHb, which responds to diverse sensory cues, including aversive and food-related odors. Optogenetic activation of BF-to-LHb circuitry drives robust, reflexive-like aversion. Furthermore, activation of this circuitry suppresses the drive to eat in a fasted state. Together, these data reveal a role of basal forebrain glutamatergic neurons in modulating LHb-associated aversion and feeding behaviors by sensing environmental cues.


Assuntos
Prosencéfalo Basal , Habenula , Habenula/fisiologia , Prosencéfalo Basal/fisiologia , Afeto , Hipotálamo/fisiologia , Comportamento Alimentar , Vias Neurais/fisiologia
3.
Nat Commun ; 10(1): 3369, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358754

RESUMO

Inhibitory interneurons are integral to sensory processing, yet revealing their cell type-specific roles in sensory circuits remains an ongoing focus. To Investigate the mouse olfactory system, we selectively remove GABAergic transmission from a subset of olfactory bulb interneurons, EPL interneurons (EPL-INs), and assay odor responses from their downstream synaptic partners - tufted cells and mitral cells. Using a combination of in vivo electrophysiological and imaging analyses, we find that inactivating this single node of inhibition leads to differential effects in magnitude, reliability, tuning width, and temporal dynamics between the two principal neurons. Furthermore, tufted and not mitral cell responses to odor mixtures become more linearly predictable without EPL-IN inhibition. Our data suggest that olfactory bulb interneurons, through exerting distinct inhibitory functions onto their different synaptic partners, play a significant role in the processing of odor information.


Assuntos
Interneurônios/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Condutos Olfatórios/fisiologia , Animais , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Inibição Neural/genética , Neurônios/citologia , Neurônios/metabolismo , Odorantes , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Olfato , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
4.
J Neurosci ; 38(6): 1443-1461, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29305536

RESUMO

The mouse olfactory bulb (OB) features continued, activity-dependent integration of adult-born neurons, providing a robust model with which to examine mechanisms of plasticity in the adult brain. We previously reported that local OB interneurons secrete the neuropeptide corticotropin-releasing hormone (CRH) in an activity-dependent manner onto adult-born granule neurons and that local CRH signaling promotes expression of synaptic machinery in the bulb. This effect is mediated via activation of the CRH receptor 1 (CRHR1), which is developmentally regulated during adult-born neuron maturation. CRHR1 is a GS-protein-coupled receptor that activates CREB-dependent transcription in the presence of CRH. Therefore, we hypothesized that locally secreted CRH activates CRHR1 to initiate circuit plasticity programs. To identify such programs, we profiled gene expression changes associated with CRHR1 activity in adult-born neurons of the OB. Here, we show that CRHR1 activity influences expression of the brain-specific Homeobox-containing transcription factor POU Class 6 Homeobox 1 (POU6f1). To elucidate the contributions of POU6f1 toward activity-dependent circuit remodeling, we targeted CRHR1+ neurons in male and female mice for cell-type-specific manipulation of POU6f1 expression. Whereas loss of POU6f1 in CRHR1+ neurons resulted in reduced dendritic complexity and decreased synaptic connectivity, overexpression of POU6f1 in CRHR1+ neurons promoted dendritic outgrowth and branching and influenced synaptic function. Together, these findings suggest that the transcriptional program directed by POU6f1 downstream of local CRH signaling in adult-born neurons influences circuit dynamics in response to activity-dependent peptide signaling in the adult brain.SIGNIFICANCE STATEMENT Elucidating mechanisms of plasticity in the adult brain is helpful for devising strategies to understand and treat neurodegeneration. Circuit plasticity in the adult mouse olfactory bulb is exemplified by both continued cell integration and synaptogenesis. We previously reported that these processes are influenced by local neuropeptide signaling in an activity-dependent manner. Here, we show that local corticotropin-releasing hormone (CRH) signaling induces dynamic gene expression changes in CRH receptor expressing adult-born neurons, including altered expression of the transcription factor POU6f1 We further show that POU6f1 is necessary for proper dendrite specification and patterning, as well as synapse development and function in adult-born neurons. Together, these findings reveal a novel mechanism by which peptide signaling modulates adult brain circuit plasticity.


Assuntos
Encéfalo/fisiologia , Plasticidade Neuronal/fisiologia , Neuropeptídeos/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Animais , Comportamento Animal/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Neurônios/ultraestrutura , Fator 3 de Transcrição de Octâmero/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Olfato/fisiologia
5.
J Neurosci ; 36(20): 5572-86, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27194336

RESUMO

UNLABELLED: Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in Methyl-CpG-binding protein 2 (MECP2). Severe breathing abnormalities are common in RTT and are reproduced in mouse models of RTT. Previously, we found that removing MeCP2 from the brainstem and spinal cord in mice caused early lethality and abnormal breathing. To determine whether loss of MeCP2 in functional components of the respiratory network causes specific breathing disorders, we used the Cre/LoxP system to differentially manipulate MeCP2 expression throughout the brainstem respiratory network, specifically within HoxA4-derived tissues, which include breathing control circuitry within the nucleus tractus solitarius and the caudal part of ventral respiratory column but do not include more rostral parts of the breathing control circuitry. To determine whether respiratory phenotypes manifested in animals with MeCP2 removed from specific pons medullary respiratory circuits, we performed whole-body plethysmography and electrophysiological recordings from in vitro brainstem slices from mice lacking MeCP2 in different circuits. Our results indicate that MeCP2 expression in the medullary respiratory network is sufficient for normal respiratory rhythm and preventing apnea. However, MeCP2 expression within components of the breathing circuitry rostral to the HoxA4 domain are neither sufficient to prevent the hyperventilation nor abnormal hypoxic ventilatory response. Surprisingly, we found that MeCP2 expression in the HoxA4 domain alone is critical for survival. Our study reveals that MeCP2 is differentially required in select respiratory components for different aspects of respiratory functions, and collectively for the integrity of this network functions to maintain proper respiration. SIGNIFICANCE STATEMENT: Breathing abnormalities are a significant clinical feature in Rett syndrome and are robustly reproduced in the mouse models of this disease. Previous work has established that alterations in the function of MeCP2, the protein encoded by the gene mutated in Rett syndrome, within the hindbrain are critical for control of normal breathing. Here we show that MeCP2 function plays distinct roles in specific brainstem regions in the genesis of various aspects of abnormal breathing. This provides insight into the pathogenesis of these breathing abnormalities in Rett syndrome, which could be used to target treatments to improve these symptoms. Furthermore, it provides further knowledge about the fundamental neural circuits that control breathing.


Assuntos
Bulbo/fisiologia , Proteína 2 de Ligação a Metil-CpG/genética , Respiração , Síndrome de Rett/fisiopatologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio , Masculino , Bulbo/metabolismo , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Síndrome de Rett/genética , Fatores de Transcrição
6.
Hum Mol Genet ; 24(9): 2662-72, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25634563

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder that is usually caused by mutations in Methyl-CpG-binding Protein 2 (MECP2). Four of the eight common disease causing mutations in MECP2 are nonsense mutations and are responsible for over 35% of all cases of RTT. A strategy to overcome disease-causing nonsense mutations is treatment with nonsense mutation suppressing drugs that allow expression of full-length proteins from mutated genes with premature in-frame stop codons. To determine if this strategy is useful in RTT, we characterized a new mouse model containing a knock-in nonsense mutation (p.R255X) in the Mecp2 locus (Mecp2(R255X)). To determine whether the truncated gene product acts as a dominant negative allele and if RTT-like phenotypes could be rescued by expression of wild-type protein, we genetically introduced an extra copy of MECP2 via an MECP2 transgene. The addition of MECP2 transgene to Mecp2(R255X) mice abolished the phenotypic abnormalities and resulted in near complete rescue. Expression of MECP2 transgene Mecp2(R255X) allele also rescued mTORC1 signaling abnormalities discovered in mice with loss of function and overexpression of Mecp2. Finally, we treated Mecp2(R255X) embryonic fibroblasts with the nonsense mutation suppressing drug gentamicin and we were able to induce expression of full-length MeCP2 from the mutant p.R255X allele. These data provide proof of concept that the p.R255X mutation of MECP2 is amenable to the nonsense suppression therapeutic strategy and provide guidelines for the extent of rescue that can be expected by re-expressing MeCP2 protein.


Assuntos
Alelos , Estudos de Associação Genética , Proteína 2 de Ligação a Metil-CpG/genética , Mutação , Fenótipo , Substituição de Aminoácidos , Animais , Comportamento Animal , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica , Gentamicinas/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transgenes
7.
J Am Heart Assoc ; 3(4)2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25142058

RESUMO

BACKGROUND: Transient receptor potential C3 (TRPC3) has been demonstrated to be involved in the regulation of vascular tone through endothelial cell (EC) hyperpolarization and endothelium-dependent hyperpolarization-mediated vasodilation. However, the mechanism by which TRPC3 regulates these processes remains unresolved. We tested the hypothesis that endothelial receptor stimulation triggers rapid TRPC3 trafficking to the plasma membrane, where it provides the source of Ca(2+) influx for small conductance calcium-activated K(+) (SKCa) channel activation and sustained EC hyperpolarization. METHODS AND RESULTS: Pressurized artery studies were performed with isolated mouse posterior cerebral artery. Treatment with a selective TRPC3 blocker (Pyr3) produced significant attenuation of endothelium-dependent hyperpolarization-mediated vasodilation and endothelial Ca(2+) response (EC-specific Ca(2+) biosensor) to intraluminal ATP. Pyr3 treatment also resulted in a reduced ATP-stimulated global Ca(2+) and Ca(2+) influx in primary cultures of cerebral endothelial cells. Patch-clamp studies with freshly isolated cerebral ECs demonstrated 2 components of EC hyperpolarization and K(+) current activation in response to ATP. The early phase was dependent on intermediate conductance calcium-activated K(+) channel activation, whereas the later sustained phase relied on SKC a channel activation. The SKC a channel-dependent phase was completely blocked with TRPC3 channel inhibition or in ECs of TRPC3 knockout mice and correlated with increased trafficking of TRPC3 (but not SKC a channel) to the plasma membrane. CONCLUSIONS: We propose that TRPC3 dynamically regulates SKC a channel activation through receptor-dependent trafficking to the plasma membrane, where it provides the source of Ca(2+) influx for sustained SKC a channel activation, EC hyperpolarization, and endothelium-dependent hyperpolarization-mediated vasodilation.


Assuntos
Cálcio/metabolismo , Células Endoteliais/metabolismo , Artéria Cerebral Posterior/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPC/genética , Vasodilatação/genética , Animais , Células Endoteliais/fisiologia , Endotélio Vascular , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Artéria Cerebral Posterior/fisiologia , Canais de Cátion TRPC/metabolismo
8.
Cardiovasc Res ; 95(4): 439-47, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22721989

RESUMO

AIMS: Microdomain signalling mechanisms underlie key aspects of artery function and the modulation of intracellular calcium, with transient receptor potential (TRP) channels playing an integral role. This study determines the distribution and role of TRP canonical type 3 (C3) channels in the control of endothelium-derived hyperpolarization (EDH)-mediated vasodilator tone in rat mesenteric artery. METHODS AND RESULTS: TRPC3 antibody specificity was verified using rat tissue, human embryonic kidney (HEK)-293 cells stably transfected with mouse TRPC3 cDNA, and TRPC3 knock-out (KO) mouse tissue using western blotting and confocal and ultrastructural immunohistochemistry. TRPC3-Pyr3 (ethyl-1-(4-(2,3,3-trichloroacrylamide)phenyl)-5-(trifluoromethyl)-1H-pyrazole-4-carboxylate) specificity was verified using patch clamp of mouse mesenteric artery endothelial and TRPC3-transfected HEK cells, and TRPC3 KO and wild-type mouse aortic endothelial cell calcium imaging and mesenteric artery pressure myography. TRPC3 distribution, expression, and role in EDH-mediated function were examined in rat mesenteric artery using immunohistochemistry and western blotting, and pressure myography and endothelial cell membrane potential recordings. In rat mesenteric artery, TRPC3 was diffusely distributed in the endothelium, with approximately five-fold higher expression at potential myoendothelial microdomain contact sites, and immunoelectron microscopy confirmed TRPC3 at these sites. Western blotting and endothelial damage confirmed primary endothelial TRPC3 expression. In rat mesenteric artery endothelial cells, Pyr3 inhibited hyperpolarization generation, and with individual SK(Ca) (apamin) or IK(Ca) (TRAM-34) block, Pyr3 abolished the residual respective IK(Ca)- and SK(Ca)-dependent EDH-mediated vasodilation. CONCLUSION: The spatial localization of TRPC3 and associated channels, receptors, and calcium stores are integral for myoendothelial microdomain function. TRPC3 facilitates endothelial SK(Ca) and IK(Ca) activation, as key components of EDH-mediated vasodilator activity and for regulating mesenteric artery tone.


Assuntos
Fatores Biológicos/metabolismo , Endotélio Vascular/metabolismo , Artérias Mesentéricas/metabolismo , Canais de Cátion TRPC/metabolismo , Vasodilatação , Animais , Pressão Arterial , Western Blotting , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/ultraestrutura , Células HEK293 , Humanos , Imuno-Histoquímica , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Masculino , Potenciais da Membrana , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/ultraestrutura , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Imunoeletrônica , Miografia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPC/efeitos dos fármacos , Canais de Cátion TRPC/genética , Transfecção , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
9.
Mol Pain ; 6: 46, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20691059

RESUMO

BACKGROUND: Transient receptor potential vanilloid 1 (TRPV1) channels are important membrane sensors on peripheral nerve endings and on supportive non-neuronal synoviocytes in the knee joint. TRPV 1 ion channels respond with activation of calcium and sodium fluxes to pH, thermal, chemical, osmotic, mechanical and other stimuli abundant in inflamed joints. In the present study, the kaolin/carrageenan (k/c) induced knee joint arthritis model in rats, as well as primary and clonal human synoviocyte cultures were used to understand the reciprocal interactions between reactive nitroxidative species (ROS) and functional TRPV1 channels. ROS generation was monitored with ROS sensitive dyes using live cell imaging in vitro and in spinal tissue histology, as well as with measurement of ROS metabolites in culture media using HPLC. RESULTS: Functional responses in the experimental arthritis model, including increased nociceptive responses (thermal and mechanical hyperalgesia and allodynia), knee joint temperature reflecting local blood flow, and spinal cord ROS elevations were reduced by the ROS scavenger PBN after intraperitoneal pretreatment. Increases in TRPV1 and ROS, generated by synoviocytes in vitro, were reciprocally blocked by TRPV1 antagonists and the ROS scavenger. Further evidence is presented that synoviocyte responses to ROS and TRPV1 activation include increases in TNFalpha and COX-2, both measured as an indicator of the inflammation in vitro. CONCLUSIONS: The results demonstrate that contributions of ROS to pronociceptive responses and neurogenic inflammation are mediated both centrally and peripherally. Responses are mediated by TRPV1 locally in the knee joint by synoviocytes, as well as by ROS-induced sensitization in the spinal cord. These findings and those of others reported in the literature indicate reciprocal interactions between TRPV1 and ROS play critical roles in the pathological and nociceptive responses active during arthritic inflammation.


Assuntos
Hiperalgesia/metabolismo , Nociceptores/metabolismo , Osteoartrite do Joelho/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Temperatura Alta , Humanos , Hiperalgesia/patologia , Cápsula Articular/metabolismo , Cápsula Articular/patologia , Nociceptores/patologia , Osteoartrite do Joelho/patologia , Ratos , Ratos Endogâmicos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Toxicol Sci ; 115(1): 1-11, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19955490

RESUMO

Xenoestrogens can affect the healthy functioning of a variety of tissues by acting as potent estrogens via nongenomic signaling pathways or by interfering with those actions of multiple physiological estrogens. Collectively, our and other studies have compared a wide range of estrogenic compounds, including some closely structurally related subgroups. The estrogens that have been studied include environmental contaminants of different subclasses, dietary estrogens, and several prominent physiological metabolites. By comparing the nongenomic signaling and functional responses to these compounds, we have begun to address the structural requirements for their actions through membrane estrogen receptors in the pituitary, in comparison to other tissues, and to gain insights into their typical non-monotonic dose-response behavior. Their multiple inputs into cellular signaling begin processes that eventually integrate at the level of mitogen-activated protein kinase activities to coordinately regulate broad cellular destinies, such as proliferation, apoptosis, or differentiation.


Assuntos
Estrogênios não Esteroides/toxicidade , Transdução de Sinais , Xenobióticos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Exposição Ambiental , Estrogênios não Esteroides/química , Estrogênios não Esteroides/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptores de Estrogênio/metabolismo , Relação Estrutura-Atividade , Xenobióticos/química , Xenobióticos/metabolismo
11.
Mol Pain ; 5: 49, 2009 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-19695100

RESUMO

BACKGROUND: We have shown functional expression of several TRP channels on human synovial cells, proposing significance in known calcium dependent proliferative and secretory responses in joint inflammation. The present study further characterizes synoviocyte TRP expression and activation responses to thermal and osmotic stimuli after pre-treatment with proinflammatory mediator tumor necrosis factor alpha (TNF-alpha, EC50 1.3221 x 10(-10) g/L). RESULTS: Fluorescent imaging of Fura-2 loaded human SW982 synoviocytes reveals immediate and delayed cytosolic calcium oscillations elicited by (1) TRPV1 agonists capsaicin and resiniferatoxin (20-40% of cells), (2) moderate and noxious temperature change, and (3) osmotic stress TRPV4 activation (11.5% of cells). TNF-alpha pre-treatment (1 ng/ml, 8-16 hr) significantly increases (doubles) capsaicin responsive cell numbers and [Ca2+]i spike frequency, as well as enhances average amplitude of temperature induced [Ca2+]i responses. With TNF-alpha pre-treatment for 8, 12, and 16 hr, activation with 36 or 45 degree bath solution induces bimodal [Ca2+]i increase (temperature controlled chamber). Initial temperature induced rapid transient spikes and subsequent slower rise reflect TRPV1 and TRPV4 channel activation, respectively. Only after prolonged TNF-alpha exposure (12 and 16 hr) is recruitment of synoviocytes observed with sensitized TRPV4 responses to hypoosmolarity (3-4 fold increase). TNF-alpha increases TRPV1 (8 hr peak) and TRPV4 (12 hr peak) immunostaining, mRNA and protein expression, with a TRPV1 shift to membrane fractions. CONCLUSION: TNF-alpha provides differentially enhanced synoviocyte TRPV1 and TRPV4 expression and [Ca2+]i response dependent on the TRP stimulus and time after exposure. Augmented relevance of TRPV1 and TRPV4 as inflammatory conditions persist would provide calcium mediated cell signaling required for pathophysiological responses of synoviocytes in inflammatory pain states.


Assuntos
Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Capsaicina/farmacologia , Linhagem Celular , Diterpenos/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Pressão Osmótica/efeitos dos fármacos , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Temperatura , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/genética
12.
Environ Health Perspect ; 117(5): 723-30, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19479013

RESUMO

BACKGROUND: Alkylphenols varying in their side-chain lengths [ethyl-, propyl-, octyl-, and nonylphenol (EP, PP, OP, and NP, respectively)] and bisphenol A (BPA) represent a large group of structurally related xenoestrogens that have endocrine-disruptive effects. Their rapid nongenomic effects that depend on structure for cell signaling and resulting functions are unknown. OBJECTIVES: We compared nongenomic estrogenic activities of alkylphenols with BPA and 17beta-estradiol (E(2)) in membrane estrogen receptor-alpha-enriched GH3/B6/F10 pituitary tumor cells. These actions included calcium (Ca) signaling, prolactin (PRL) release, extracellular-regulated kinase (ERK) phosphorylation, and cell proliferation. METHODS: We imaged Ca using fura-2, measured PRL release via radioimmunoassay, detected ERK phosphorylation by fixed cell immunoassay, and estimated cell number using the crystal violet assay. RESULTS: All compounds caused increases in Ca oscillation frequency and intracellular Ca volume at 100 fM to 1 nM concentrations, although long-chain alkylphenols were most effective. All estrogens caused rapid PRL release at concentrations as low as 1 fM to 10 pM; the potency of EP, PP, and NP exceeded that of E(2). All compounds at 1 nM produced similar increases in ERK phosphorylation, causing rapid peaks at 2.5-5 min, followed by inactivation and additional 60-min peaks (except for BPA). Dose-response patterns of ERK activation at 5 min were similar for E2, BPA, and PP, whereas EP caused larger effects. Only E2 and NP increased cell number. Some rapid estrogenic responses showed correlations with the hydrophobicity of estrogenic molecules; the more hydrophobic OP and NP were superior at Ca and cell proliferation responses, whereas the less hydrophobic EP and PP were better at ERK activations. CONCLUSIONS: Alkylphenols are potent estrogens in evoking these nongenomic responses contributing to complex functions; their hydrophobicity can largely predict these behaviors.


Assuntos
Fenóis/química , Fenóis/farmacologia , Animais , Compostos Benzidrílicos , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Estradiol/farmacologia , Fosforilação/efeitos dos fármacos , Neoplasias Hipofisárias , Prolactina/metabolismo , Radioimunoensaio , Ratos
13.
J Mol Signal ; 4: 2, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19400946

RESUMO

BACKGROUND: Estradiol (E2) mediates various intracellular signaling cascades from the plasma membrane via several estrogen receptors (ERs). The pituitary is an estrogen-responsive tissue, and we have previously reported that E2 can activate mitogen-activated protein kinases (MAPKs) such as ERK1/2 and JNK1/2/3 in the membrane ERalpha (mERalpha)-enriched GH3/B6/F10 rat pituitary tumor cell line. Phytoestrogens are compounds found in plants and foods such as soybeans, alfalfa sprouts, and red grapes. They are structurally similar to E2 and share a similar mechanism of action through their binding to ERs. Phytoestrogens bind to nuclear ERs with a much lower affinity and therefore are less potent in mediating genomic responses. However, little is known about their ability to act via mERs to mediate nongenomic effects. METHODS: To investigate the activation of different nongenomic pathways, and determine the involvement of mERalpha, we measured prolactin (PRL) release by radio-immunoassay, MAPK activations (ERK1/2 and JNK1/2/3) via a quantitative plate immunoassay, and intracellular [Ca2+] by Fura-2 fluorescence imaging in cells treated with E2 or four different phytoestrogens (coumestrol, daidzein, genistein, and trans-resveratrol). RESULTS: Coumesterol and daidzein increased PRL release similar to E2 in GH3/B6/F10 cells, while genistein and trans-resveratrol had no effect. All of these compounds except genistein activated ERK1/2 signaling at 1-10 picomolar concentrations; JNK 1/2/3 was activated by all compounds at a 100 nanomolar concentration. All compounds also caused rapid Ca2+ uptake, though in unique dose-dependent Ca2+ response patterns for several aspects of this response. A subclone of GH3 cells expressing low levels of mERalpha (GH3/B6/D9) did not respond to any phytoestrogen treatments for any of these responses, suggesting that these nongenomic effects were mediated via mERalpha. CONCLUSION: Phytoestrogens were much more potent in mediating these nongenomic responses (activation of MAPKs, PRL release, and increased intracellular [Ca2+]) via mERalpha than was previously reported for genomic responses. The unique non-monotonic dose responses and variant signaling patterns caused by E2 and all tested phytoestrogens suggest that complex and multiple signaling pathways or binding partners could be involved. By activating these different nongenomic signaling pathways, phytoestrogens could have significant physiological consequences for pituitary cell functions.

14.
FASEB J ; 22(9): 3328-36, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18541692

RESUMO

Physiological estrogens, including estrone (E(1)), estradiol (E(2)), and estriol (E(3)), fluctuate with life stage, suggesting specific roles for them in biological and disease processes. We compared their nongenomic signaling and functional actions in GH3/B6/F10 rat pituitary tumor cells. All hormones caused prolactin release at 1 min; the lowest effective concentrations were 10(-11) M E(2), 10(-10) M E(1), and 10(-7) M E(3). All estrogens increased the oscillation frequency of calcium (Ca) spikes, with the same time delay (approximately 200 s) at all levels (10(-15) to 10(-9) M). At some concentrations, E(1) and E(3) provoked more Ca-responding cells than E(2). The amplitude and volume of Ca peaks were elevated by all hormones at > or = 10(-15) M. All hormones caused cell proliferation, with the lowest effective concentrations of E(2) (10(-15) M) > E(1) (10(-12) M) > E(3) (10(-10) M); E(2) caused higher maximal cell numbers at most concentrations. All estrogens caused oscillating extracellular-regulated kinase (ERK) activations, with relative potencies of E(1) and E(2) > E(3). All estrogens were ineffective in activation of ERKs or causing proliferation in a subline expressing low levels of membrane estrogen receptor-alpha. Dose-response patterns were frequently nonmonotonic. Therefore, the hormones E(1) and E(3), which have been designated "weak" estrogens in genomic actions, are strong estrogens in the nongenomic signaling pathways and functional responses in the pituitary.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Estradiol/fisiologia , Estriol/fisiologia , Estrona/fisiologia , Neoplasias Hipofisárias/fisiopatologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Prolactina/metabolismo , Ratos
15.
Am J Physiol Cell Physiol ; 291(3): C424-32, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16597917

RESUMO

The transient receptor potential (TRP) channels are important membrane sensors, responding to thermal, chemical, osmotic, or mechanical stimuli by activation of calcium and sodium fluxes. In this study, three distinct TRP channels were detected and their role established in mediating cytosolic free calcium concentration ([Ca(2+)](cyt)) response in tumor-derived SW982 synoviocytes and primary cultures of human synovial cells from patients with inflammatory arthropathies. As shown by fura-2 ratio measurements while cells were incubated in a temperature-regulated chamber, significant [Ca(2+)](cyt) elevation was elicited by rapid changes in bath temperature, application of TRPV1 receptor agonists capsaicin and resiniferatoxin, or a cold receptor stimulator, icilin. Temperature thresholds for calcium response were determined to be 12 +/- 1 degrees C for cold and 28 +/- 2 degrees C for heat activation. Temperature increases or decreases beyond these thresholds resulted in a significant rise in the magnitude of [Ca(2+)](cyt) spikes. Observed changes in [Ca(2+)](cyt) were completely abolished in calcium-free medium and thus resulted from direct calcium entry through TRP channels rather then by activation of voltage-dependent calcium channels. Two heat sensitive channels, TRPV1 and TRPV4, and a cold-sensitive channel, TRPA1, were detected by RT-PCR. Minimal mRNA for TRPV3 or TRPM8 was amplified. The RT-PCR results support the data obtained with the [Ca(2+)](cyt) measurements. We propose that the TRP channels are functionally expressed in human synoviocytes and may play a critical role in adaptive or pathological changes in articular surfaces during arthritic inflammation.


Assuntos
Cálcio/metabolismo , Membrana Sinovial/citologia , Canais de Potencial de Receptor Transitório/metabolismo , Artrite Reumatoide/fisiopatologia , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Capsaicina/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Condrocalcinose/fisiopatologia , Citosol/metabolismo , Diterpenos/farmacologia , Temperatura Alta , Humanos , Inflamação , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Pirimidinonas/farmacologia , RNA Mensageiro , Membrana Sinovial/fisiologia , Canal de Cátion TRPA1 , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...