Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Toxins (Basel) ; 14(10)2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36287977

RESUMO

Acid-sensing ion channels (ASICs) are voltage-independent H+-gated cation channels largely expressed in the nervous system of rodents and humans. At least six isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) associate into homotrimers or heterotrimers to form functional channels with highly pH-dependent gating properties. This review provides an update on the pharmacological profiles of animal peptide toxins targeting ASICs, including PcTx1 from tarantula and related spider toxins, APETx2 and APETx-like peptides from sea anemone, and mambalgin from snake, as well as the dimeric protein snake toxin MitTx that have all been instrumental to understanding the structure and the pH-dependent gating of rodent and human cloned ASICs and to study the physiological and pathological roles of native ASICs in vitro and in vivo. ASICs are expressed all along the pain pathways and the pharmacological data clearly support a role for these channels in pain. ASIC-targeting peptide toxins interfere with ASIC gating by complex and pH-dependent mechanisms sometimes leading to opposite effects. However, these dual pH-dependent effects of ASIC-inhibiting toxins (PcTx1, mambalgin and APETx2) are fully compatible with, and even support, their analgesic effects in vivo, both in the central and the peripheral nervous system, as well as potential effects in humans.


Assuntos
Canais Iônicos Sensíveis a Ácido , Venenos de Aranha , Animais , Humanos , Roedores/metabolismo , Venenos de Aranha/química , Peptídeos/química , Analgésicos/farmacologia , Dor/tratamento farmacológico
2.
Eur J Pharmacol ; 934: 175296, 2022 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-36162458

RESUMO

AIM: This study aimed to investigate the promoting effect of acid-sensing ion channel 1a (ASIC1a) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and its mechanisms. METHODS: In this experiment, the ALI rat model was induced by intratracheal injection of LPS, and the ASIC1a specific blocker psalmotoxin-1 (PcTx-1) was injected into the tail vein before LPS administration once. Western blot, immunofluorescence, immunohistochemistry and real-time PCR methods were used to detect ASIC1a and apoptosis-related proteins expressions in lung tissue and RLE-6TN rat type II alveolar epithelial cells. Confocal Laser Scanning Microscopy was used to detect Ca2+ fluorescence intensity in RLE-6TN cells. RESULTS: PcTx-1 pretreatment not only inhibited the pathological changes of LPS-induced ALI in lung tissue, but also inhibited lung dysfunction. PcTx-1 also reduced the increased levels of the apoptosis-related proteins B-cell lymphoma-2-associated X (Bax) and cleaved cysteinyl aspartate specific proteinase 3 (Cleaved caspase-3) and increased the decreased level of B-cell lymphoma-2 (Bcl-2) in the lung tissue of the model group. LPS-induced changes in mitochondrial membrane potential and calcium influx in alveolar epithelial cells were also reversed by PcTx-1. CONCLUSION: ASIC1a induces an apoptotic response in ALI through mitochondrial apoptosis.


Assuntos
Canais Iônicos Sensíveis a Ácido , Lesão Pulmonar Aguda , Animais , Ratos , Canais Iônicos Sensíveis a Ácido/genética , Canais Iônicos Sensíveis a Ácido/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Ácido Aspártico , Proteína X Associada a bcl-2/metabolismo , Cálcio/metabolismo , Caspase 3/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Mieloblastina/metabolismo
3.
Elife ; 102021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34319232

RESUMO

ASIC1a is a proton-gated sodium channel involved in modulation of pain, fear, addiction, and ischemia-induced neuronal injury. We report isolation and characterization of alpaca-derived nanobodies (Nbs) that specifically target human ASIC1a. Cryo-electron microscopy of the human ASIC1a channel at pH 7.4 in complex with one of these, Nb.C1, yielded a structure at 2.9 Å resolution. It is revealed that Nb.C1 binds to a site overlapping with that of the Texas coral snake toxin (MitTx1) and the black mamba venom Mambalgin-1; however, the Nb.C1-binding site does not overlap with that of the inhibitory tarantula toxin psalmotoxin-1 (PcTx1). Fusion of Nb.C1 with PcTx1 in a single polypeptide markedly enhances the potency of PcTx1, whereas competition of Nb.C1 and MitTx1 for binding reduces channel activation by the toxin. Thus, Nb.C1 is a molecular tool for biochemical and structural studies of hASIC1a; a potential antidote to the pain-inducing component of coral snake bite; and a candidate to potentiate PcTx1-mediated inhibition of hASIC1a in vivo for therapeutic applications.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Anticorpos de Domínio Único/química , Canais Iônicos Sensíveis a Ácido/ultraestrutura , Animais , Camelídeos Americanos , Microscopia Crioeletrônica , Ligação Proteica , Anticorpos de Domínio Único/ultraestrutura
4.
Transl Cancer Res ; 9(12): 7519-7530, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35117352

RESUMO

BACKGROUND: The microenvironment of various tumor tissues is acidic. Acid-sensing ion channels (ASICs) are a class of ligand-gated ion channels which are sensitive to extracellular protons and are often highly expressed in tumor tissues. Breast cancer, whose extracellular microenvironment is thought to be acidic, is the most common cancer type among females in the world. METHODS: Thirty breast cancer tissues and adjacent normal tissues of patients were collected from 2009 to 2015 at the Xinhua hospital affiliated to Shanghai Jiao Tong University School of Medicine. The expression of acid-sensing ion channel 1a (ASIC1a), a subtype of ASICs family, was detected by immunohistochemistry in breast cancer tissues, and the effect of ASIC1a on the physiological activity of tumor cells was analyzed in vitro and in vivo experiments. RESULTS: In this study, it was found that ASIC1a is highly expressed in the tissues of breast cancer patients. In vitro experiments revealed that down-regulation of ASIC1a by its antagonist PcTx-1 or ASIC1a siRNA could significantly weaken the migration, proliferation and invasion of tumor cells. In vivo studies, down-regulation or inhibition of the ASIC1a could inhibit breast tumor growth. CONCLUSIONS: The high expression of ASIC1a might be related to the enhanced biological activity of breast cancer cells. Whether ASIC1a is a potential therapeutic target for some types of breast cancer deserves further study.

5.
Biochem Pharmacol ; 163: 381-390, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30849303

RESUMO

Acid-sensing ion channels (ASICs) are primary acid sensors in the mammalian nervous system that are activated by protons under conditions of local acidosis. They have been implicated in a range of pathologies including ischemic stroke (ASIC1a subtype) and peripheral pain (ASIC1b and ASIC3). Although the spider venom peptide PcTx1 is the best-studied ASIC modulator and is neuroprotective in rodent models of ischemic stroke, little experimental work has been done to examine its molecular interaction with human ASIC1a or the off-target ASIC1b. The complementary face of the acidic pocket binding site of PcTx1 is where these channels differ in sequence. We show here that although PcTx1 is 10-fold less potent at human ASIC1a than the rat channel, the apparent affinity for the two channels is comparable. We examined the pharmacophore of PcTx1 for human ASIC1a and rat ASIC1b, and show that inhibitory and stimulatory effects at each ASIC1 variant is driven mostly by a shared set of core peptide pharmacophore residues that bind to the thumb domain, while peptide residues that interact with the complementary face of the biding site underlie species and subtype-dependent differences in activity that may allow manipulation of ASIC1 variant selectivity. Finally, the stimulatory effect of PcTx1 on rat ASIC1a when applied under mildly alkaline pH correlates with low receptor occupancy. These new insights into the interactions between PcTx1 with ASIC1 subtypes demonstrates the complexity of its mechanism of action, and highlights important implications to consider when using PcTx1 as a pharmacological tool to study ASIC function.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Peptídeos/metabolismo , Venenos de Aranha/metabolismo , Canais Iônicos Sensíveis a Ácido/química , Animais , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Humanos , Modelos Moleculares , Mutação , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Peptídeos/química , Ligação Proteica , Conformação Proteica , Engenharia de Proteínas , Subunidades Proteicas , Ratos , Especificidade da Espécie , Venenos de Aranha/química , Xenopus laevis
6.
Neuropharmacology ; 125: 429-440, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28802647

RESUMO

Acid-Sensing Ion Channels (ASICs) are cation channels activated by extracellular acidification that emerge as potential pharmacological targets in pain and other neurological disorders. Here, we compare the pharmacological modulation of ASIC1a and ASIC3 channels by amiloride and 2-guanidine-4-methylquinazoline (GMQ), two compounds commonly used for their in vitro and in vivo investigation. We analyzed the effect of amiloride on the pH-dependent activation and inactivation, the relative influence of the extracellular domain and the transmembrane/cytosolic domains on the effect of amiloride and GMQ using chimeras between ASIC1a and ASIC3, and how these compounds potentiate the physiologically relevant ASIC3 sustained window current. We showed that amiloride and GMQ shift the pH-dependent activation and inactivation in the same directions, which depend on the channel, and that their effects rely on the nature of the extracellular domain but can be indirectly modulated in their amplitude by the transmembrane/cytosolic domains. The extracellular domain explains the pharmacological potentiating effect of amiloride and GMQ on the window current in ASIC3, and why these compounds failed to generate a window current in ASIC1a. Amiloride and GMQ have similar and purely additive effects suggesting that they act through a common unique binding site different from acidic pockets. Finally, a simple cycle analysis using GMQ that targets the nonproton ligand-sensor, and two peptide inhibitors of ASIC1a targeting the acidic pockets (PcTx1 and mambalgin-1), shows overlap between the mechanisms by which GMQ and PcTx1 modify inactivation and suggests shared mechanisms of regulation of the pH-dependent inactivation of ASIC1a between these two regions.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Amilorida/farmacologia , Guanidinas/farmacologia , Quinazolinas/farmacologia , Animais , Venenos Elapídicos/farmacologia , Concentração de Íons de Hidrogênio , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Oócitos , Peptídeos/farmacologia , Domínios Proteicos , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Venenos de Aranha/farmacologia , Xenopus
7.
Sci Adv ; 2(12): e1600516, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28028533

RESUMO

Glioma patients often suffer from epileptic seizures because of the tumor's impact on the brain physiology. Using the rat glioma cell line C6 as a model system, we performed long-term live recordings of the electrical activity of glioma populations in an ultrasensitive detection method. The transducer exploits large-area electrodes that maximize double-layer capacitance, thus increasing the sensitivity. This strategy allowed us to record glioma electrical activity. We show that although glioma cells are nonelectrogenic, they display a remarkable electrical burst activity in time. The low-frequency current noise after cell adhesion is dominated by the flow of Na+ ions through voltage-gated ion channels. However, after an incubation period of many hours, the current noise markedly increased. This electric bursting phenomenon was not associated with apoptosis because the cells were viable and proliferative during the period of increased electric activity. We detected a rapid cell culture medium acidification accompanying this event. By using specific inhibitors, we showed that the electrical bursting activity was prompted by extracellular pH changes, which enhanced Na+ ion flux through the psalmotoxin 1-sensitive acid-sensing ion channels. Our model of pH-triggered bursting was unambiguously supported by deliberate, external acidification of the cell culture medium. This unexpected, acidosis-driven electrical activity is likely to directly perturb, in vivo, the functionality of the healthy neuronal network in the vicinity of the tumor bulk and may contribute to seizures in glioma patients.


Assuntos
Fenômenos Eletrofisiológicos , Glioma/fisiopatologia , Concentração de Íons de Hidrogênio , Neurônios/citologia , Animais , Linhagem Celular Tumoral , Humanos , Rede Nervosa , Ratos , Canais de Sódio/fisiologia
8.
F1000Res ; 5: 1822, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28105306

RESUMO

Tissue loss after spinal trauma is biphasic, with initial mechanical/haemorrhagic damage at the time of impact being followed by gradual secondary expansion into adjacent, previously unaffected tissue. Limiting the extent of this secondary expansion of tissue damage has the potential to preserve greater residual spinal cord function in patients. The acute tissue hypoxia resulting from spinal cord injury (SCI) activates acid-sensing ion channel 1a (ASIC1a). We surmised that antagonism of this channel should provide neuroprotection and functional preservation after SCI. We show that systemic administration of the spider-venom peptide PcTx1, a selective inhibitor of ASIC1a, improves locomotor function in adult Sprague Dawley rats after thoracic SCI. The degree of functional improvement correlated with the degree of tissue preservation in descending white matter tracts involved in hind limb locomotor function. Transcriptomic analysis suggests that PcTx1-induced preservation of spinal cord tissue does not result from a reduction in apoptosis, with no evidence of down-regulation of key genes involved in either the intrinsic or extrinsic apoptotic pathways. We also demonstrate that trauma-induced disruption of blood-spinal cord barrier function persists for at least 4 days post-injury for compounds up to 10 kDa in size, whereas barrier function is restored for larger molecules within a few hours. This temporary loss of barrier function provides a " treatment window" through which systemically administered drugs have unrestricted access to spinal tissue in and around the sites of trauma. Taken together, our data provide evidence to support the use of ASIC1a inhibitors as a therapeutic treatment for SCI. This study also emphasizes the importance of objectively grading the functional severity of initial injuries (even when using standardized impacts) and we describe a simple scoring system based on hind limb function that could be adopted in future studies.

9.
Neuropharmacology ; 99: 650-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26320544

RESUMO

Acid-sensing ion channel 1a (ASIC1a) is the primary acid sensor in mammalian brain and plays a major role in neuronal injury following cerebral ischemia. Evidence that inhibition of ASIC1a might be neuroprotective following stroke was previously obtained using "PcTx1 venom" from the tarantula Psalmopeous cambridgei. We show here that the ASIC1a-selective blocker PcTx1 is present at only 0.4% abundance in this venom, leading to uncertainty as to whether the observed neuroprotective effects were due to PcTx1 blockade of ASIC1a or inhibition of other ion channels and receptors by the hundreds of peptides and small molecules present in the venom. We therefore examined whether pure PcTx1 is neuroprotective in a conscious model of stroke via direct inhibition of ASIC1a. A focal reperfusion model of stroke was induced in conscious spontaneously hypertensive rats (SHR) by administering endothelin-1 to the middle cerebral artery via a surgically implanted cannula. Two hours later, SHR were treated with a single intracerebroventricular (i.c.v.) dose of PcTx1 (1 ng/kg), an ASIC1a-inactive mutant of PcTx1 (1 ng/kg), or saline, and ledged beam and neurological tests were used to assess the severity of symptomatic changes. PcTx1 markedly reduced cortical and striatal infarct volumes measured 72 h post-stroke, which correlated with improvements in neurological score, motor function and preservation of neuronal architecture. In contrast, the inactive PcTx1 analogue had no effect on stroke outcome. This is the first demonstration that selective pharmacological inhibition of ASIC1a is neuroprotective in conscious SHRs, thus validating inhibition of ASIC1a as a potential treatment for stroke.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Venenos de Aranha/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Canais Iônicos Sensíveis a Ácido/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Endotelina-1 , Infarto da Artéria Cerebral Média , Masculino , Atividade Motora/efeitos dos fármacos , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Peptídeos/genética , Ratos Endogâmicos SHR , Índice de Gravidade de Doença , Venenos de Aranha/genética , Aranhas , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
10.
Proc Natl Acad Sci U S A ; 111(24): 8961-6, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24889629

RESUMO

Stimulating presynaptic terminals can increase the proton concentration in synapses. Potential receptors for protons are acid-sensing ion channels (ASICs), Na(+)- and Ca(2+)-permeable channels that are activated by extracellular acidosis. Those observations suggest that protons might be a neurotransmitter. We found that presynaptic stimulation transiently reduced extracellular pH in the amygdala. The protons activated ASICs in lateral amygdala pyramidal neurons, generating excitatory postsynaptic currents. Moreover, both protons and ASICs were required for synaptic plasticity in lateral amygdala neurons. The results identify protons as a neurotransmitter, and they establish ASICs as the postsynaptic receptor. They also indicate that protons and ASICs are a neurotransmitter/receptor pair critical for amygdala-dependent learning and memory.


Assuntos
Canais Iônicos Sensíveis a Ácido/genética , Tonsila do Cerebelo/fisiologia , Plasticidade Neuronal , Neurotransmissores/metabolismo , Prótons , Sinapses/fisiologia , Bloqueadores do Canal Iônico Sensível a Ácido/química , Acidose , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Eletrodos , Potenciais Pós-Sinápticos Excitadores , Concentração de Íons de Hidrogênio , Canais Iônicos/química , Aprendizagem , Potenciação de Longa Duração , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Peptídeos/química , Venenos de Aranha/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA