Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurosci ; 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35882560

RESUMO

A rod-shaped appendage called a primary cilium projects from the soma of most central neurons in the mammalian brain. The importance of cilia within the nervous system is highlighted by the fact that human syndromes linked to primary cilia dysfunction, collectively termed ciliopathies, are associated with numerous neuropathologies, including hyperphagia-induced obesity, neuropsychiatric disorders, and learning and memory deficits. Neuronal cilia are enriched with signaling molecules, including specific G protein-coupled receptors (GPCRs) and their downstream effectors, suggesting they act as sensory organelles that respond to neuromodulators in the extracellular space. We previously showed that GPCR ciliary localization is disrupted in neurons from mouse models of the ciliopathy Bardet-Biedl syndrome (BBS). Based on this finding we hypothesized that mislocalization of ciliary GPCRs may impact receptor signaling and contribute to the BBS phenotypes. Here, we show that disrupting localization of the ciliary GPCR dopamine receptor 1 (D1) in male and female mice, either by loss of a BBS protein or loss of the cilium itself, specifically in D1-expressing neurons, results in obesity. Interestingly, the weight gain is associated with reduced locomotor activity, rather than increased food intake. Moreover, loss of a BBS protein or cilia on D1-expressing neurons leads to a reduction in D1-mediated signaling. Together, these results indicate that cilia impact D1 activity in the nervous system and underscore the importance of neuronal cilia for proper GPCR signaling.SIGNIFICANCE STATEMENT:Most mammalian neurons possess solitary appendages called primary cilia. These rod-shaped structures are enriched with signaling proteins, such as G protein-coupled receptors (GPCRs), suggesting they respond to neuromodulators. This study examines the consequences of disrupting ciliary localization of the GPCR dopamine receptor 1 (D1) in D1-expressing neurons. Remarkably, mice that have either abnormal accumulation of D1 in cilia or loss of D1 ciliary localization become obese. In both cases the obesity is associated with lower locomotor activity rather than overeating. As D1 activation increases locomotor activity, these results are consistent with a reduction in D1 signaling. Indeed, we found that D1-mediated signaling is reduced in brain slices from both mouse models. Thus, cilia impact D1 signaling in the brain.

2.
Proc Natl Acad Sci U S A ; 113(42): E6496-E6505, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27688759

RESUMO

Oligodendrocytes have recently been implicated in the pathophysiology of amyotrophic lateral sclerosis (ALS). Here we show that, in vitro, mutant superoxide dismutase 1 (SOD1) mouse oligodendrocytes induce WT motor neuron (MN) hyperexcitability and death. Moreover, we efficiently derived human oligodendrocytes from a large number of controls and patients with sporadic and familial ALS, using two different reprogramming methods. All ALS oligodendrocyte lines induced MN death through conditioned medium (CM) and in coculture. CM-mediated MN death was associated with decreased lactate production and release, whereas toxicity in coculture was lactate-independent, demonstrating that MN survival is mediated not only by soluble factors. Remarkably, human SOD1 shRNA treatment resulted in MN rescue in both mouse and human cultures when knockdown was achieved in progenitor cells, whereas it was ineffective in differentiated oligodendrocytes. In fact, early SOD1 knockdown rescued lactate impairment and cell toxicity in all lines tested, with the exclusion of samples carrying chromosome 9 ORF 72 (C9orf72) repeat expansions. These did not respond to SOD1 knockdown nor did they show lactate release impairment. Our data indicate that SOD1 is directly or indirectly involved in ALS oligodendrocyte pathology and suggest that in this cell type, some damage might be irreversible. In addition, we demonstrate that patients with C9ORF72 represent an independent patient group that might not respond to the same treatment.


Assuntos
Esclerose Lateral Amiotrófica/genética , Neurônios Motores/metabolismo , Oligodendroglia/metabolismo , Superóxido Dismutase-1/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Apoptose , Biomarcadores , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Comunicação Celular , Morte Celular , Diferenciação Celular , Sobrevivência Celular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Ácido Láctico/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Superóxido Dismutase-1/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(28): 10335-40, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982149

RESUMO

Most central neurons in the mammalian brain possess an appendage called a primary cilium that projects from the soma into the extracellular space. The importance of these organelles is highlighted by the fact that primary cilia dysfunction is associated with numerous neuropathologies, including hyperphagia-induced obesity, hypogonadism, and learning and memory deficits. Neuronal cilia are enriched for signaling molecules, including certain G protein-coupled receptors (GPCRs), suggesting that neuronal cilia sense and respond to neuromodulators in the extracellular space. However, the impact of cilia on signaling to central neurons has never been demonstrated. Here, we show that the kisspeptin receptor (Kiss1r), a GPCR that is activated by kisspeptin to regulate the onset of puberty and adult reproductive function, is enriched in cilia projecting from mouse gonadotropin-releasing hormone (GnRH) neurons. Interestingly, GnRH neurons in adult animals are multiciliated and the percentage of GnRH neurons possessing multiple Kiss1r-positive cilia increases during postnatal development in a progression that correlates with sexual maturation. Remarkably, disruption of cilia selectively on GnRH neurons leads to a significant reduction in kisspeptin-mediated GnRH neuronal activity. To our knowledge, this result is the first demonstration of cilia disruption affecting central neuronal activity and highlights the importance of cilia for proper GPCR signaling.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Cílios/genética , Cílios/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Maturidade Sexual/fisiologia
4.
J Neurosci ; 31(26): 9723-34, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21715637

RESUMO

Acid-sensing ion channel (ASIC) subunits associate to form homomeric or heteromeric proton-gated ion channels in neurons throughout the nervous system. The ASIC1a subunit plays an important role in establishing the kinetics of proton-gated currents in the CNS, and activation of ASIC1a homomeric channels induces neuronal death after local acidosis that accompanies cerebral ischemia. The ASIC2b subunit is expressed in the brain in a pattern that overlaps ASIC1a, yet the contribution of ASIC2b has remained elusive. We find that coexpression of ASIC2b with ASIC1a in Xenopus oocytes results in novel proton-gated currents with properties distinct from ASIC1a homomeric channels. In particular, ASIC2b/1a heteromeric channels are inhibited by the nonselective potassium channel blockers tetraethylammonium and barium. In addition, steady-state desensitization is induced at more basic pH values, and Big Dynorphin sensitivity is enhanced in these unique heteromeric channels. Cultured hippocampal neurons show proton-gated currents consistent with ASIC2b contribution, and these currents are lacking in neurons from mice with an ACCN1 (ASIC2) gene disruption. Finally, we find that these ASIC2b/1a heteromeric channels contribute to acidosis-induced neuronal death. Together, our results show that ASIC2b confers unique properties to heteromeric channels in central neurons. Furthermore, these data indicate that ASIC2, like ASIC1, plays a role in acidosis-induced neuronal death and implicate the ASIC2b/1a subtype as a novel pharmacological target to prevent neuronal injury after stroke.


Assuntos
Acidose/metabolismo , Morte Celular/fisiologia , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Acidose/fisiopatologia , Análise de Variância , Animais , Eletrofisiologia , Hipocampo/fisiopatologia , Camundongos , Xenopus
5.
Am J Physiol Cell Physiol ; 303(7): C699-710, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22843794

RESUMO

The acid-sensing ion channels (ASICs) are a family of proton-sensing channels expressed throughout the nervous system. Their activity is linked to a variety of complex behaviors including fear, anxiety, pain, depression, learning, and memory. ASICs have also been implicated in neuronal degeneration accompanying ischemia and multiple sclerosis. As a whole, ASICs represent novel therapeutic targets for several clinically important disorders. An understanding of the correlation between ASIC structure and function will help to elucidate their mechanism of action and identify potential therapeutics that specifically target these ion channels. Despite the seemingly simple nature of proton binding, multiple studies have shown that proton-dependent gating of ASICs is quite complex, leading to activation and desensitization through distinct structural components. This review will focus on the structural aspects of ASIC gating in response to both protons and the newly discovered activators GMQ and MitTx. ASIC modulatory compounds and their action on proton-dependent gating will also be discussed. This review is dedicated to the memory of Dale Benos, who made a substantial contribution to our understanding of ASIC activity.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Canais Iônicos Sensíveis a Ácido/fisiologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Animais , Humanos , Poro Nuclear/química , Poro Nuclear/metabolismo , Poro Nuclear/fisiologia , Relação Estrutura-Atividade
6.
Cell Mol Life Sci ; 68(17): 2951-60, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21152952

RESUMO

Primary cilia are nearly ubiquitous cellular appendages that provide important sensory and signaling functions. Ciliary dysfunction underlies numerous human diseases, collectively termed ciliopathies. Primary cilia have distinct functions on different cell types and these functions are defined by the signaling proteins that localize to the ciliary membrane. Neurons throughout the mammalian brain possess primary cilia upon which certain G protein-coupled receptors localize. Yet, the precise signaling proteins present on the vast majority of neuronal cilia are unknown. Here, we report that dopamine receptor 1 (D1) localizes to cilia on mouse central neurons, thereby implicating neuronal cilia in dopamine signaling. Interestingly, ciliary localization of D1 is dynamic, and the receptor rapidly translocates to and from cilia in response to environmental cues. Notably, the translocation of D1 from cilia requires proteins mutated in the ciliopathy Bardet-Biedl syndrome (BBS), and we find that one of the BBS proteins, Bbs5, specifically interacts with D1.


Assuntos
Proteínas de Transporte/metabolismo , Cílios/metabolismo , Receptores de Dopamina D1/metabolismo , Animais , Síndrome de Bardet-Biedl/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas do Citoesqueleto , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Proteínas de Ligação a Fosfato , Proteínas/metabolismo , Receptores de Dopamina D1/análise , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
7.
Front Pain Res (Lausanne) ; 3: 894651, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35812017

RESUMO

Low back pain is a leading cause of disability worldwide and studies have demonstrated intervertebral disc (IVD) degeneration as a major risk factor. While many in vitro models have been developed and used to study IVD pathophysiology and therapeutic strategies, the etiology of IVD degeneration is a complex multifactorial process involving crosstalk of nearby tissues and systemic effects. Thus, the use of appropriate in vivo models is necessary to fully understand the associated molecular, structural, and functional changes and how they relate to pain. Mouse models have been widely adopted due to accessibility and ease of genetic manipulation compared to other animal models. Despite their small size, mice lumbar discs demonstrate significant similarities to the human IVD in terms of geometry, structure, and mechanical properties. While several different mouse models of IVD degeneration exist, greater standardization of the methods for inducing degeneration and the development of a consistent set of output measurements could allow mouse models to become a stronger tool for clinical translation. This article reviews current mouse models of IVD degeneration in the context of clinical translation and highlights a critical set of output measurements for studying disease pathology or screening regenerative therapies with an emphasis on pain phenotyping. First, we summarized and categorized these models into genetic, age-related, and mechanically induced. Then, the outcome parameters assessed in these models are compared including, molecular, cellular, functional/structural, and pain assessments for both evoked and spontaneous pain. These comparisons highlight a set of potential key parameters that can be used to validate the model and inform its utility to screen potential therapies for IVD degeneration and their translation to the human condition. As treatment of symptomatic pain is important, this review provides an emphasis on critical pain-like behavior assessments in mice and explores current behavioral assessments relevant to discogenic back pain. Overall, the specific research question was determined to be essential to identify the relevant model with histological staining, imaging, extracellular matrix composition, mechanics, and pain as critical parameters for assessing degeneration and regenerative strategies.

8.
J Biol Chem ; 285(53): 41852-62, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21036899

RESUMO

The acid-sensing ion channels (ASICs) are proton-gated cation channels activated when extracellular pH declines. In rodents, the Accn2 gene encodes transcript variants ASIC1a and ASIC1b, which differ in the first third of the protein and display distinct channel properties. In humans, ACCN2 transcript variant 2 (hVariant 2) is homologous to mouse ASIC1a. In this article, we study two other human ACCN2 transcript variants. Human ACCN2 transcript variant 1 (hVariant 1) is not present in rodents and contains an additional 46 amino acids directly preceding the proposed channel gate. We report that hVariant 1 does not produce proton-gated currents under normal conditions when expressed in heterologous systems. We also describe a third human ACCN2 transcript variant (hVariant 3) that is similar to rodent ASIC1b. hVariant 3 is more abundantly expressed in dorsal root ganglion compared with brain and shows basic channel properties analogous to rodent ASIC1b. Yet, proton-gated currents from hVariant 3 are significantly more permeable to calcium than either hVariant 2 or rodent ASIC1b, which shows negligible calcium permeability. hVariant 3 also displays a small acid-dependent sustained current. Such a sustained current is particularly intriguing as ASIC1b is thought to play a role in sensory transduction in rodents. In human DRG neurons, hVariant 3 could induce sustained calcium influx in response to acidic pH and make a major contribution to acid-dependent sensations, such as pain.


Assuntos
Cálcio/química , Proteínas do Tecido Nervoso/química , Canais de Sódio/química , Canais Iônicos Sensíveis a Ácido , Sequência de Aminoácidos , Animais , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , Humanos , Concentração de Íons de Hidrogênio , Íons/química , Camundongos , Dados de Sequência Molecular , Neurônios/metabolismo , Oócitos/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Xenopus laevis/metabolismo
9.
Proc Natl Acad Sci U S A ; 105(11): 4242-6, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18334641

RESUMO

Primary cilia are ubiquitous cellular appendages that provide important yet not well understood sensory and signaling functions. Ciliary dysfunction underlies numerous human genetic disorders. However, the precise defects in cilia function and the basis of disease pathophysiology remain unclear. Here, we report that the proteins disrupted in the human ciliary disorder Bardet-Biedl syndrome (BBS) are required for the localization of G protein-coupled receptors to primary cilia on central neurons. We demonstrate a lack of ciliary localization of somatostatin receptor type 3 (Sstr3) and melanin-concentrating hormone receptor 1 (Mchr1) in neurons from mice lacking the Bbs2 or Bbs4 gene. Because Mchr1 is involved in the regulation of feeding behavior and BBS is associated with hyperphagia-induced obesity, our results suggest that altered signaling caused by mislocalization of ciliary signaling proteins underlies the BBS phenotypes. Our results also provide a potential molecular mechanism to link cilia defects with obesity.


Assuntos
Síndrome de Bardet-Biedl/metabolismo , Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Síndrome de Bardet-Biedl/genética , Células Cultivadas , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Proteínas/genética
10.
J Neurosci ; 29(45): 14371-80, 2009 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19906984

RESUMO

Acid-sensing ion channel 1a (ASIC1a) promotes neuronal damage during pathological acidosis. ASIC1a undergoes a process called steady-state desensitization in which incremental pH reductions desensitize the channel and prevent activation when the threshold for acid-dependent activation is reached. We find that dynorphin A and big dynorphin limit steady-state desensitization of ASIC1a and acid-activated currents in cortical neurons. Dynorphin potentiation of ASIC1a activity is independent of opioid or bradykinin receptor activation but is prevented in the presence of PcTx1, a peptide which is known to bind the extracellular domain of ASIC1a. This suggests that dynorphins interact directly with ASIC1a to enhance channel activity. Inducing steady-state desensitization prevents ASIC1a-mediated cell death during prolonged acidosis. This neuroprotection is abolished in the presence of dynorphins. Together, these results define ASIC1a as a new nonopioid target for dynorphin action and suggest that dynorphins enhance neuronal damage following ischemia by preventing steady-state desensitization of ASIC1a.


Assuntos
Acidose/fisiopatologia , Dinorfinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Análise de Variância , Animais , Morte Celular/fisiologia , Células Cultivadas , Hipocampo/fisiopatologia , Concentração de Íons de Hidrogênio , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Peptídeos , Prótons , Receptores da Bradicinina/metabolismo , Receptores Opioides/metabolismo , Canais de Sódio/genética , Venenos de Aranha/farmacologia , Xenopus laevis
11.
Neurobiol Stress ; 13: 100240, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33344696

RESUMO

Gulf War illness is associated with a combination of exposure to war-related chemical agents and traumatic stress. Currently, there are no effective treatments, and the pathophysiology remains elusive. Neurological problems are among the most commonly reported symptoms. In this study, we investigated the glutamatergic system in the hippocampi of mice exposed to war-related chemical agents and stress. Mice developed Gulf War illness-like symptoms, including mood deficits, cognitive impairments, and fatigue. They exhibited the following pathological changes in hippocampi: elevated extracellular glutamate levels, impaired glutamatergic synapses, astrocyte atrophy, loss of interneurons, and decreased neurogenesis. LDN/OSU-215111 is a small-molecule that can strengthen the structure and function of both the astrocytic processes and the glutamatergic synapses that together form the tripartite synapses. We found that LDN/OSU-215111 effectively prevented the development of mood and cognitive deficits in mice when treatment was implemented immediately following the exposure. Moreover, when symptoms were already present, LDN/OSU-215111 still significantly ameliorated these deficits; impressively, benefits were sustained one month after treatment cessation, indicating disease modification. LDN/OSU-215111 effectively normalized hippocampal pathological changes. Overall, this study provides strong evidence that restoration of tripartite glutamatergic synapses by LDN/OSU-215111 is a potential therapy for Gulf War illness.

12.
Alzheimers Res Ther ; 11(1): 75, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31439023

RESUMO

BACKGROUND: The lack of effective treatment options for Alzheimer's disease (AD) is of momentous societal concern. Synaptic loss is the hallmark of AD that correlates best with impaired memory and occurs early in the disease process, before the onset of clinical symptoms. We have developed a small-molecule, pyridazine-based series that enhances the structure and function of both the glial processes and the synaptic boutons that form the tripartite synapse. Previously, we have shown that these pyridazine derivatives exhibit profound efficacy in an amyloid precursor protein AD model. Here, we evaluated the efficacy of an advanced compound, LDN/OSU-0215111, in rTg4510 mice-an aggressive tauopathy model. METHODS: rTg4510 mice were treated orally with vehicle or LDN/OSU-0215111 (10 mg/kg) daily from the early symptomatic stage (2 months old) to moderate (4 months old) and severe (8 months old) disease stages. At each time point, mice were subjected to a battery of behavioral tests to assess the activity levels and cognition. Also, tissue collections were performed on a subset of mice to analyze the tripartite synaptic changes, neurodegeneration, gliosis, and tau phosphorylation as assessed by immunohistochemistry and Western blotting. At 8 months of age, a subset of rTg4510 mice treated with compound was switched to vehicle treatment and analyzed behaviorally and biochemically 30 days after treatment cessation. RESULTS: At both the moderate and severe disease stages, compound treatment normalized cognition and behavior as well as reduced synaptic loss, neurodegeneration, tau hyperphosporylation, and neuroinflammation. Importantly, after 30 days of treatment cessation, the benefits of compound treatment were sustained, indicating disease modification. We also found that compound treatment rapidly and robustly reduced tau hyperphosphorylation/deposition possibly via the inhibition of GSK3ß. CONCLUSIONS: The results show that LDN/OSU-0215111 provides benefits for multiple aspects of tauopathy-dependent pathology found in Alzheimer's disease including tripartite synapse normalization and reduction of toxic tau burden, which, in turn, likely accounted for normalized cognition and activity levels in compound-treated rTg4510 mice. This study, in combination with our previous work regarding the benefit of pyridazine derivatives against amyloid-dependent pathology, strongly supports pyridazine derivatives as a viable, clinically relevant, and disease-modifying treatment for many of the facets of Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Transportador 2 de Aminoácido Excitatório , Piridazinas/farmacologia , Sinapses/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Piridazinas/administração & dosagem , Piridazinas/análise , Sinapses/patologia
13.
Neuron ; 34(3): 463-77, 2002 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11988176

RESUMO

Many central neurons possess large acid-activated currents, yet their molecular identity is unknown. We found that eliminating the acid sensing ion channel (ASIC) abolished H(+)-gated currents in hippocampal neurons. Neuronal H(+)-gated currents and transient acidification are proposed to play a role in synaptic transmission. Investigating this possibility, we found ASIC in hippocampus, in synaptosomes, and in dendrites localized at synapses. Moreover, loss of ASIC impaired hippocampal long-term potentiation. ASIC null mice had reduced excitatory postsynaptic potentials and NMDA receptor activation during high-frequency stimulation. Consistent with these findings, null mice displayed defective spatial learning and eyeblink conditioning. These results identify ASIC as a key component of acid-activated currents and implicate these currents in processes underlying synaptic plasticity, learning, and memory.


Assuntos
Aprendizagem/fisiologia , Proteínas de Membrana , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Proteínas/metabolismo , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Animais , Condicionamento Palpebral , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores , Hipocampo/citologia , Hipocampo/metabolismo , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Ácido Cinurênico/farmacologia , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Canais de Sódio/genética , Transmissão Sináptica/fisiologia
14.
Neuroscience ; 388: 224-238, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30056115

RESUMO

Excitatory amino acid transporter 2 (EAAT2) is primarily located in perisynaptic astrocytic processes (PAP) where it plays a critical role in synaptic glutamate homeostasis. Dysregulation of EAAT2 at the translational level has been implicated in a myriad of neurological diseases. We previously discovered that pyridazine analogs can activate EAAT2 translation. Here, we sought to further refine the site and mechanism of compound action. We found that in vivo, compound treatment increased EAAT2 expression only in the PAP of astrocytes where EAAT2 mRNA also was identified. Direct application of compound to isolated PAP induced de novo EAAT2 protein synthesis, indicating that compound activates translation locally in the PAP. Using a screening process, we identified a set of PAP proteins that are rapidly up-regulated following compound treatment and a subset of these PAP proteins may be locally synthesized in the PAP. Importantly, these identified proteins are associated with the structural and functional capacity of the PAP, indicating compound enhanced plasticity of the PAP. Concomitantly, we found that pyridazine analogs increase synaptic protein expression in the synapse and enhance hippocampal long-term potentiation. This was not dependent upon compound-mediated local translation in neurons. This suggests that compound enhances the structural and functional capacity of the PAP which in turn facilitates enhanced plasticity of the tripartite synapse. Overall, this provides insight into the mechanism action site of pyridazine derivatives as well as the growing appreciation of the dynamic regulation and functional aspects of the PAP at the tripartite synapse.


Assuntos
Astrócitos/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Piridazinas/farmacologia , Sinapses/efeitos dos fármacos , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Prosencéfalo/citologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Proteoma/efeitos dos fármacos , RNA Mensageiro/metabolismo , Sinapses/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Técnicas de Cultura de Tecidos
15.
Exp Neurol ; 303: 1-11, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29407729

RESUMO

Membrane potential (VM) depolarization occurs immediately following cerebral ischemia and is devastating for the astrocyte homeostasis and neuronal signaling. Previously, an excessive release of extracellular K+ and glutamate has been shown to underlie an ischemia-induced VM depolarization. Ischemic insults should impair membrane ion channels and disrupt the physiological ion gradients. However, their respective contribution to ischemia-induced neuronal and glial depolarization and loss of neuronal excitability are unanswered questions. A short-term oxygen-glucose deprivation (OGD) was used for the purpose of examining the acute effect of ischemic conditions on ion channel activity and physiological K+ gradient in neurons and glial cells. We show that a 30 min OGD treatment exerted no measurable damage to the function of membrane ion channels in neurons, astrocytes, and NG2 glia. As a result of the resilience of membrane ion channels, neuronal spikes last twice as long as our previously reported 15 min time window. In the electrophysiological analysis, a 30 min OGD-induced dissipation of transmembrane K+ gradient contributed differently in brain cell depolarization: severe in astrocytes and neurons, and undetectable in NG2 glia. The discrete cellular responses to OGD corresponded to a total loss of 69% of the intracellular K+ contents in hippocampal slices as measured by Inductively Coupled Plasma Mass Spectrometry (ICP-MS). A major brain cell depolarization mechanism identified here is important for our understanding of cerebral ischemia pathology. Additionally, further understanding of the resilient response of NG2 glia to ischemia-induced intracellular K+ loss and depolarization should facilitate the development of future stroke therapy.


Assuntos
Astrócitos/fisiologia , Fenômenos Biofísicos/fisiologia , Glucose/metabolismo , Hipóxia/fisiopatologia , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Potássio/metabolismo , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Fenômenos Biofísicos/efeitos dos fármacos , Condutividade Elétrica , Feminino , Células Gigantes/fisiologia , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigênio/farmacologia , Técnicas de Patch-Clamp , Proteoglicanas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
16.
J Biotechnol ; 122(4): 393-411, 2006 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-16309774

RESUMO

A new field of gene transfer is emerging as a simple, effective means to drive the expression foreign genes in cells: ultrasound-mediated gene transfer or sonoporation. We report here that sonoporation is an effective means of gene transfer for cultured neurons, a cell type that has been difficult to transfect. Neuronal cell types that are effectively sonoporated include chick retinal neurons, chick dorsal forebrain, chick optic tectum, PC12 cells, rat cerebellar neurons and mouse hippocampal neurons. Depending on the type of cell and conditions of sonoporation the transfection efficacy was as high as 20%. Sonoporation of plasmid DNA was effective for cells adherent to a substrate and for free-floating cells that were freshly dissociated. In the free-floating preparations, between 60 and 95% of the cells that were transfected were neuronal, as much as 90% higher than that observed for other methods of gene transfer including adenovirus and lipid-based transfection methods. We conclude that sonoporation is a simple, effective and inexpensive means by which to preferentially transfect DNA into neuronal cells.


Assuntos
Eletroporação/métodos , Técnicas de Transferência de Genes , Neurônios/metabolismo , Transfecção/métodos , Ultrassom , Animais , Morte Celular , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Embrião de Galinha , Galinhas/metabolismo , Relação Dose-Resposta a Droga , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Camundongos , Neurônios/citologia , Plasmídeos/administração & dosagem , Plasmídeos/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Ratos , Retina/citologia , Retina/metabolismo
17.
J Neurosci ; 23(13): 5496-502, 2003 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-12843249

RESUMO

The acid-sensing ion channel, ASIC1, contributes to synaptic plasticity in the hippocampus and to hippocampus-dependent spatial memory. To explore the role of ASIC1 in brain, we examined the distribution of ASIC1 protein. Surprisingly, although ASIC1 was present in the hippocampal circuit, it was much more abundant in several areas outside the hippocampus. ASIC1 was enriched in areas with strong excitatory synaptic input such as the glomerulus of the olfactory bulb, whisker barrel cortex, cingulate cortex, striatum, nucleus accumbens, amygdala, and cerebellar cortex. Because ASIC1 levels were particularly high in the amygdala, we focused further on this area. We found that extracellular acidosis elicited a greater current density in amygdala neurons than hippocampal neurons and that disrupting the ASIC1 gene eliminated H+-evoked currents in the amygdala. We also tested the effect of ASIC1 on amygdala-dependent behavior; ASIC1-null mice displayed deficits in cue and context fear conditioning, yet baseline fear on the elevated plus maze was intact. These studies suggest that ASIC1 is distributed to regions supporting high levels of synaptic plasticity and contributes to the neural mechanisms of fear conditioning.


Assuntos
Encéfalo/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Proteínas de Membrana , Proteínas do Tecido Nervoso , Canais de Sódio/metabolismo , Sinapses/metabolismo , Canais Iônicos Sensíveis a Ácido , Estimulação Acústica , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Animais , Comportamento Animal/fisiologia , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Canais de Sódio/deficiência , Canais de Sódio/genética
18.
Neuropharmacology ; 94: 36-41, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25592215

RESUMO

The acid sensing ion channels (ASICs) are proton-gated cation channels expressed throughout the nervous system. ASICs are activated during acidic pH fluctuations, and recent work suggests that they are involved in excitatory synaptic transmission. ASICs can also induce neuronal degeneration and death during pathological extracellular acidosis caused by ischemia, autoimmune inflammation, and traumatic injury. Many endogenous neuromodulators target ASICs to affect their biophysical characteristics and contributions to neuronal activity. One of the most unconventional types of modulation occurs with the interaction of ASICs and neuropeptides. Collectively, FMRFamide-related peptides and dynorphins potentiate ASIC activity by decreasing the proton-sensitivity of steady state desensitization independent of G protein-coupled receptor activation. By decreasing the proton-sensitivity of steady state desensitization, the FMRFamide-related peptides and dynorphins permit ASICs to remain active at more acidic basal pH. Unlike the dynorphins, some FMRFamide-related peptides also potentiate ASIC activity by slowing inactivation and increasing the sustained current. Through mechanistic studies, the modulation of ASICs by FMRFamide-related peptides and dynorphins appears to be through distinct interactions with the extracellular domain of ASICs. Dynorphins are expressed throughout the nervous system and can increase neuronal death during prolonged extracellular acidosis, suggesting that the interaction between dynorphins and ASICs may have important consequences for the prevention of neurological injury. The overlap in expression of FMRFamide-related peptides with ASICs in the dorsal horn of the spinal cord suggests that their interaction may have important consequences for the treatment of pain during injury and inflammation. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Neuropeptídeos/metabolismo , Animais , Humanos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo
19.
PLoS One ; 8(8): e71733, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977127

RESUMO

Acid-sensing ion channel 1a (ASIC1a) is a proton-gated cation channel that contributes to fear and pain as well as neuronal damage following persistent cerebral acidosis. Neuropeptides can affect acid-induced neuronal injury by altering ASIC1a inactivation and/or steady-state desensitization. Yet, exactly how ASIC1a inactivation and desensitization occur or are modulated by peptides is not completely understood. We found that regions of the extracellular palm domain and the ß(11-12) linker are important for inactivation and steady-state desensitization of ASIC1a. The single amino acid substitutions L280C and L415C dramatically enhanced the rate of inactivation and altered the pH-dependence of steady-state desensitization. Further, the use of methanethiosulfonate (MTS) reagents suggests that the lower palm region (L280C) undergoes a conformational change when ASIC1a transitions from closed to desensitized. We determined that L280C also displays an altered response to the RFamide peptide, FRRFamide. Further, the presence of FRRFamide limited MTS modification of L280C. Together, these results indicate a potential role of the lower palm domain in peptide modulation and suggest RFamide-related peptides promote conformational changes within this region. These data provide empirical support for the idea that L280, and likely this region of the central vestibule, is intimately involved in channel inactivation and desensitization.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Canais Iônicos Sensíveis a Ácido/metabolismo , Ativação do Canal Iônico , Neuropeptídeos/metabolismo , Animais , Humanos , Mesilatos/metabolismo , Mutação , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Xenopus laevis
20.
J Neurophysiol ; 99(2): 426-41, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18094106

RESUMO

Acid-sensing ion channels (ASICs) are H(+)-gated channels that produce transient cation currents in response to extracellular acid. ASICs are expressed in neurons throughout the brain, and ASIC1 knockout mice show behavioral impairments in learning and memory. The role of ASICs in synaptic transmission, however, is not thoroughly understood. We analyzed the involvement of ASICs in synaptic transmission using microisland cultures of hippocampal neurons from wild-type and ASIC knockout mice. There was no significant difference in single action potential (AP)-evoked excitatory postsynaptic currents (EPSCs) between wild-type and ASIC knockout neurons. However, paired-pulse ratios (PPRs) were reduced and spontaneous miniature EPSCs (mEPSCs) occurred at a higher frequency in ASIC1 knockout neurons compared with wild-type neurons. The progressive block of NMDA receptors by an open channel blocker, MK-801, was also faster in ASIC1 knockout neurons. The amplitude and decay time constant of mEPSCs, as well as the size and refilling of the readily releasable pool, were similar in ASIC1 knockout and wild-type neurons. Finally, the release probability, which was estimated directly as the ratio of AP-evoked to hypertonic sucrose-induced charge transfer, was increased in ASIC1 knockout neurons. Transfection of ASIC1a into ASIC1 knockout neurons increased the PPRs, suggesting that alterations in release probability were not the result of developmental compensation within the ASIC1 knockout mice. Together, these findings demonstrate that neurons from ASIC1 knockout mice have an increased probability of neurotransmitter release and indicate that ASIC1a can affect presynaptic mechanisms of synaptic transmission.


Assuntos
Hipocampo/citologia , Proteínas de Membrana/deficiência , Proteínas do Tecido Nervoso/deficiência , Neurônios/citologia , Terminações Pré-Sinápticas/metabolismo , Probabilidade , Canais de Sódio/deficiência , Canais Iônicos Sensíveis a Ácido , Amilorida/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Interações Medicamentosas , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Prótons , Bloqueadores dos Canais de Sódio/farmacologia , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA