Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Nat Rev Neurosci ; 23(10): 596-610, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35831443

RESUMO

The perception of nociceptive signals, which are translated into pain, plays a fundamental role in the survival of organisms. Because pain is linked to a negative sensation, animals learn to avoid noxious signals. These signals are detected by receptors, which include some members of the transient receptor potential (TRP) family of ion channels that act as transducers of exogenous and endogenous noxious cues. These proteins have been in the focus of the field of physiology for several years, and much knowledge of how they regulate the function of the cell types and organs where they are expressed has been acquired. The last decade has been especially exciting because the 'resolution revolution' has allowed us to learn the molecular intimacies of TRP channels using cryogenic electron microscopy. These findings, in combination with functional studies, have provided insights into the role played by these channels in the generation and maintenance of pain.


Assuntos
Canais de Potencial de Receptor Transitório , Animais , Dor , Sensação/fisiologia , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
2.
Neurochem Res ; 49(3): 684-691, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38017313

RESUMO

In the spinal cord, attenuation of the inhibitory action of glycine is related to an increase in both inflammatory and diabetic neuropathic pain; however, the glycine receptor involvement in diabetic neuropathy has not been reported. We determined the expression of the glycine receptor subunits (α1-α3 and ß) in streptozotocin-induced diabetic Long-Evans rats by qPCR and Western blot. The total mRNA and protein expression (whole spinal cord homogenate) of the α1, α3, and ß subunits did not change during diabetes; however, the α2 subunit mRNA, but not the protein, was overexpressed 45 days after diabetes induction. By contrast, the synaptic expression of the α1 and α2 subunits decreased in all the studied stages of diabetes, but that of the α3 subunit increased on day 45 after diabetes induction. Intradermal capsaicin produced higher paw-licking behavior in the streptozotocin-induced diabetic rats than in the control animals. In addition, the nocifensive response was higher at 45 days than at 20 days. During diabetes, the expression of the glycine receptor was altered in the spinal cord, which strongly suggests its involvement in diabetic neuropathy.


Assuntos
Diabetes Mellitus Experimental , Neuropatias Diabéticas , Ratos , Animais , Glicina/metabolismo , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Estreptozocina/toxicidade , Neuropatias Diabéticas/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Ratos Long-Evans , Medula Espinal/metabolismo , RNA Mensageiro/metabolismo
3.
J Neurosci ; 42(7): 1173-1183, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-34965978

RESUMO

The physical interaction and functional cross talk among the different subtypes of neuronal nicotinic acetylcholine receptors (nAChRs) expressed in the various tissues is unknown. Here, we have investigated this issue between the only two nAChRs subtypes expressed, the α7 and α3ß4 subtypes, in a human native neuroendocrine cell (the chromaffin cell) using electrophysiological patch-clamp, fluorescence, and Förster resonance energy transfer (FRET) techniques. Our data show that α7 and α3ß4 receptor subtypes require their mutual and maximal efficacy of activation to increase their expression, to avoid their desensitization, and therefore, to increase their activity. In this way, after repetitive stimulation with acetylcholine (ACh), α7 and α3ß4 receptor subtypes do not desensitize, but they do with choline. The nicotinic current increase associated with the α3ß4 subtype is dependent on Ca2+ In addition, both receptor subtypes physically interact. Interaction and expression of both subtypes are reversibly reduced by tyrosine and serine/threonine phosphatases inhibition, not by Ca2+ In addition, expression is greater in human chromaffin cells from men compared to women, but FRET efficiency is not affected. Together, our findings indicate that human α7 and α3ß4 subtypes mutually modulate their expression and activity, providing a promising line of research to pharmacologically regulate their activity.SIGNIFICANCE STATEMENT Desensitization of nicotinic receptors is accepted to occur with repetitive agonist stimulation. However, here we show that human native α3ß4 and α7 nicotinic acetylcholine receptor (nAChR) subtypes do not desensitize, and instead, increase their activity when they are activated by the physiological agonist acetylcholine (ACh). An indispensable requirement is the activation of the other receptor subtype with maximal efficacy, and the presence of Ca2+ to cooperate in the case of the α3ß4 current increase. Because choline is an α3ß4 partial agonist, it will act as a limiting factor of nicotinic currents enhancement in the absence of ACh, but in its presence, it will further potentiate α7 currents.


Assuntos
Células Cromafins/metabolismo , Receptor Cross-Talk/fisiologia , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL
4.
Pflugers Arch ; 475(5): 595-606, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36964781

RESUMO

The primary function of dystrophin is to form a link between the cytoskeleton and the extracellular matrix. In addition to this crucial structural function, dystrophin also plays an essential role in clustering and organizing several signaling proteins, including ion channels. Proteomic analysis of the whole rodent brain has stressed the role of some components of the dystrophin-associated glycoprotein complex (DGC) as potential interacting proteins of the voltage-gated Ca2+ channels of the CaV2 subfamily. The interaction of CaV2 with signaling and scaffolding proteins, such as the DGC components, may influence their function, stability, and location in neurons. This work aims to study the interaction between dystrophin and CaV2.1. Our immunoprecipitation data showed the presence of a complex formed by CaV2.1, CaVα2δ-1, CaVß4e, Dp140, and α1-syntrophin in the brain. Furthermore, proximity ligation assays (PLA) showed that CaV2.1 and CaVα2δ-1 interact with dystrophin in the hippocampus and cerebellum. Notably, Dp140 and α1-syntrophin increase CaV2.1 protein stability, half-life, permanence in the plasma membrane, and current density through recombinant CaV2.1 channels. Therefore, we have identified the Dp140 and α1-syntrophin as novel interaction partners of CaV2.1 channels in the mammalian brain. Consistent with previous findings, our work provides evidence of the role of DGC in anchoring and clustering CaV channels in a macromolecular complex.


Assuntos
Distrofina , Proteômica , Animais , Distrofina/genética , Distrofina/metabolismo , Mamíferos/metabolismo , Neurônios/metabolismo
5.
Adv Exp Med Biol ; 1422: 245-277, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36988884

RESUMO

Transient receptor potential (TRP) ion channels are proteins that are expressed by diverse tissues and that play pivotal functions in physiology. These channels are polymodal and are activated by several stimuli. Among TRPs, some members of this family of channels respond to changes in ambient temperature and are known as thermoTRPs. These proteins respond to heat or cold in the noxious range and some of them to temperatures considered innocuous, as well as to mechanical, osmotic, and/or chemical stimuli. In addition to this already complex ability to respond to different signals, the activity of these ion channels can be fine-tuned by lipids. Two lipids well known to modulate ion channel activity are phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol. These lipids can either influence the function of these proteins through direct interaction by binding to a site in the structure of the ion channel or through indirect mechanisms, which can include modifying membrane properties, such as curvature and rigidity, by regulating their expression or by modulating the actions of other molecules or signaling pathways that affect the physiology of ion channels. Here, we summarize the key aspects of the regulation of thermoTRP channels by PIP2 and cholesterol.


Assuntos
Canais de Potencial de Receptor Transitório , Canais de Potencial de Receptor Transitório/metabolismo , Temperatura , Temperatura Baixa , Fosfatidilinositóis , Colesterol/metabolismo
6.
Int J Mol Sci ; 24(20)2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37894963

RESUMO

There is a clear need to expand the toolkit of adequate mouse models and cell lines available for preclinical studies of high-grade neuroendocrine lung carcinoma (small cell lung carcinoma (SCLC) and large cell neuroendocrine carcinoma (LCNEC)). SCLC and LCNEC are two highly aggressive tumor types with dismal prognoses and few therapeutic options. Currently, there is an extreme paucity of material, particularly in the case of LCNEC. Given the lack of murine cell lines and transplant models of LCNEC, the need is imperative. In this study, we generated and examined new models of LCNEC and SCLC transplantable cell lines derived from our previously developed primary mouse LCNEC and SCLC tumors. RNA-seq analysis demonstrated that our cell lines and syngeneic tumors maintained the transcriptome program from the original transgenic primary tumor and displayed strong similarities to human SCLC or LCNEC. Importantly, the SCLC transplanted cell lines showed the ability to metastasize and mimic this characteristic of the human condition. In summary, we generated mouse cell line tools that allow further basic and translational research as well as preclinical testing of new treatment strategies for SCLC and LCNEC. These tools retain important features of their human counterparts and address the lack of LCNEC disease models.


Assuntos
Carcinoma de Células Grandes , Carcinoma Neuroendócrino , Carcinoma de Células Pequenas , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Animais , Camundongos , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Células Pequenas/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/patologia , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/patologia , Pulmão/patologia
7.
Gastroenterology ; 161(1): 301-317.e16, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33819485

RESUMO

BACKGROUND & AIMS: Limited understanding of pruritus mechanisms in cholestatic liver diseases hinders development of antipruritic treatments. Previous studies implicated lysophosphatidic acid (LPA) as a potential mediator of cholestatic pruritus. METHODS: Pruritogenicity of lysophosphatidylcholine (LPC), LPA's precursor, was examined in naïve mice, cholestatic mice, and nonhuman primates. LPC's pruritogenicity involving keratinocyte TRPV4 was studied using genetic and pharmacologic approaches, cultured keratinocytes, ion channel physiology, and structural computational modeling. Activation of pruriceptor sensory neurons by microRNA-146a (miR-146a), secreted from keratinocytes, was identified by in vitro and ex vivo Ca2+ imaging assays. Sera from patients with primary biliary cholangitis were used for measuring the levels of LPC and miR-146a. RESULTS: LPC was robustly pruritic in mice. TRPV4 in skin keratinocytes was essential for LPC-induced itch and itch in mice with cholestasis. Three-dimensional structural modeling, site-directed mutagenesis, and channel function analysis suggested a TRPV4 C-terminal motif for LPC binding and channel activation. In keratinocytes, TRPV4 activation by LPC induced extracellular release of miR-146a, which activated TRPV1+ sensory neurons to cause itch. LPC and miR-146a levels were both elevated in sera of patients with primary biliary cholangitis with itch and correlated with itch intensity. Moreover, LPC and miR-146a were also increased in sera of cholestatic mice and elicited itch in nonhuman primates. CONCLUSIONS: We identified LPC as a novel cholestatic pruritogen that induces itch through epithelia-sensory neuron cross talk, whereby it directly activates skin keratinocyte TRPV4, which rapidly releases miR-146a to activate skin-innervating TRPV1+ pruriceptor sensory neurons. Our findings support the new concept of the skin, as a sensory organ, playing a critical role in cholestatic itch, beyond liver, peripheral sensory neurons, and central neural pathways supporting pruriception.


Assuntos
Colestase/complicações , Queratinócitos/metabolismo , Lisofosfatidilcolinas , Prurido/metabolismo , Células Receptoras Sensoriais/metabolismo , Pele/inervação , Canais de Cátion TRPV/metabolismo , Adulto , Idoso , Animais , Comportamento Animal , Células Cultivadas , Colestase/genética , Colestase/metabolismo , Colestase/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Prurido/induzido quimicamente , Prurido/genética , Prurido/fisiopatologia , Transdução de Sinais , Canais de Cátion TRPV/genética
8.
Proc Natl Acad Sci U S A ; 116(44): 22300-22306, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31611390

RESUMO

High-grade neuroendocrine lung malignancies (large-cell neuroendocrine cell carcinoma, LCNEC, and small-cell lung carcinoma, SCLC) are among the most deadly lung cancer conditions with no optimal clinical management. The biological relationships between SCLC and LCNEC are still largely unknown and a current matter of debate as growing molecular data reveal high heterogeneity with potential therapeutic consequences. Here we describe murine models of high-grade neuroendocrine lung carcinomas generated by the loss of 4 tumor suppressors. In an Rbl1-null background, deletion of Rb1, Pten, and Trp53 floxed alleles after Ad-CMVcre infection in a wide variety of lung epithelial cells produces LCNEC. Meanwhile, inactivation of these genes using Ad-K5cre in basal cells leads to the development of SCLC, thus differentially influencing the lung cancer type developed. So far, a defined model of LCNEC has not been reported. Molecular and transcriptomic analyses of both models revealed strong similarities to their human counterparts. In addition, a 68Ga-DOTATOC-based molecular-imaging method provides a tool for detection and monitoring the progression of the cancer. These data offer insight into the biology of SCLC and LCNEC, providing a useful framework for development of compounds and preclinical investigations in accurate immunocompetent models.


Assuntos
Carcinoma de Células Pequenas/genética , Genes Supressores de Tumor , Neoplasias Pulmonares/genética , Tumores Neuroendócrinos/genética , Animais , Carcinoma de Células Pequenas/diagnóstico por imagem , Carcinoma de Células Pequenas/patologia , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Camundongos , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/patologia , Octreotida/análogos & derivados , Compostos Organometálicos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Proteínas de Ligação a Retinoblastoma/genética , Proteínas de Ligação a Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Transcriptoma , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(7): E1657-E1666, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29378958

RESUMO

The Transient Receptor Potential Vanilloid 1 (TRPV1) ion channel is expressed in nociceptors where, when activated by chemical or thermal stimuli, it functions as an important transducer of painful and itch-related stimuli. Although the interaction of TRPV1 with proteins that regulate its function has been previously explored, their modulation by chaperones has not been elucidated, as is the case for other mammalian TRP channels. Here we show that TRPV1 physically interacts with the Sigma 1 Receptor (Sig-1R), a chaperone that binds progesterone, an antagonist of Sig-1R and an important neurosteroid associated to the modulation of pain. Antagonism of Sig-1R by progesterone results in the down-regulation of TRPV1 expression in the plasma membrane of sensory neurons and, consequently, a decrease in capsaicin-induced nociceptive responses. This is observed both in males treated with a synthetic antagonist of Sig-1R and in pregnant females where progesterone levels are elevated. This constitutes a previously undescribed mechanism by which TRPV1-dependent nociception and pain can be regulated.


Assuntos
Dor/metabolismo , Receptores sigma/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/metabolismo , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dor/genética , Progesterona/metabolismo , Ligação Proteica , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPV/genética , Receptor Sigma-1
10.
Int J Mol Sci ; 21(10)2020 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-32408609

RESUMO

The Transient Receptor Potential Vanilloid 1 (TRPV1) channel is a polymodal protein with functions widely linked to the generation of pain. Several agonists of exogenous and endogenous nature have been described for this ion channel. Nonetheless, detailed mechanisms and description of binding sites have been resolved only for a few endogenous agonists. This review focuses on summarizing discoveries made in this particular field of study and highlighting the fact that studying the molecular details of activation of the channel by different agonists can shed light on biophysical traits that had not been previously demonstrated.


Assuntos
Ativação do Canal Iônico , Domínios Proteicos , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Animais , Sítios de Ligação/genética , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , Canais de Cátion TRPV/genética
11.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255148

RESUMO

The Transient Receptor Vanilloid 1 (TRPV1) or capsaicin receptor is a nonselective cation channel, which is abundantly expressed in nociceptors. This channel is an important transducer of several noxious stimuli, having a pivotal role in pain development. Several TRPV1 studies have focused on understanding its structure and function, as well as on the identification of compounds that regulate its activity. The intracellular roles of these channels have also been explored, highlighting TRPV1's actions in the homeostasis of Ca2+ in organelles such as the mitochondria. These studies have evidenced how the activation of TRPV1 affects mitochondrial functions and how this organelle can regulate TRPV1-mediated nociception. The close relationship between this channel and mitochondria has been determined in neuronal and non-neuronal cells, demonstrating that TRPV1 activation strongly impacts on cell physiology. This review focuses on describing experimental evidence showing that TRPV1 influences mitochondrial function.


Assuntos
Sinalização do Cálcio/genética , Mitocôndrias/genética , Dor/genética , Canais de Cátion TRPV/genética , Animais , Cálcio/metabolismo , Humanos , Mitocôndrias/metabolismo , Nociceptividade/fisiologia , Dor/fisiopatologia , Transdução de Sinais/genética
12.
Int J Mol Sci ; 21(11)2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32481620

RESUMO

Transient Receptor Potential (TRP) channels are a family of ion channels whose members are distributed among all kinds of animals, from invertebrates to vertebrates. The importance of these molecules is exemplified by the variety of physiological roles they play. Perhaps, the most extensively studied member of this family is the TRPV1 ion channel; nonetheless, the activity of TRPV4 has been associated to several physio and pathophysiological processes, and its dysfunction can lead to severe consequences. Several lines of evidence derived from animal models and even clinical trials in humans highlight TRPV4 as a therapeutic target and as a protein that will receive even more attention in the near future, as will be reviewed here.


Assuntos
Canais de Cátion TRPV/fisiologia , Animais , Cálcio/metabolismo , Bovinos , Endotélio Vascular/metabolismo , Humanos , Rim/metabolismo , Camundongos , Microcirculação , Dor/metabolismo , Permeabilidade , Prognóstico , Domínios Proteicos , Ratos , Vasos Retinianos , Pele/metabolismo
13.
Int J Mol Sci ; 21(11)2020 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471309

RESUMO

Transient receptor potential (TRP) channels are remarkable transmembrane protein complexes that are essential for the physiology of the tissues in which they are expressed. They function as non-selective cation channels allowing for the signal transduction of several chemical, physical and thermal stimuli and modifying cell function. These channels play pivotal roles in the nervous and reproductive systems, kidney, pancreas, lung, bone, intestine, among others. TRP channels are finely modulated by different mechanisms: regulation of their function and/or by control of their expression or cellular/subcellular localization. These mechanisms are subject to being affected by several endogenously-produced compounds, some of which are of a lipidic nature such as steroids. Fascinatingly, steroids and TRP channels closely interplay to modulate several physiological events. Certain TRP channels are affected by the typical genomic long-term effects of steroids but others are also targets for non-genomic actions of some steroids that act as direct ligands of these receptors, as will be reviewed here.


Assuntos
Androgênios/metabolismo , Estrogênios/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos , Canais de Potencial de Receptor Transitório/química , Canais de Potencial de Receptor Transitório/genética
14.
J Neurochem ; 148(3): 359-372, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30347483

RESUMO

High catecolamine plasma levels because of sympathetic nervous system over-activity contribute to cirrhosis progression. The aim of this study was to investigate whether chromaffin cells of the adrenal gland might potentiate the deleterious effect exerted by this over-activity. Electrophysiological patch-clamp and amperometric experiments with carbon-fibre electrodes were conducted in single chromaffin cells of control and CCl4 -induced cirrhotic rats. The spontaneous action potential firing frequency was increased in chromaffin cells of cirrhotic rats with respect to control rats. The exocytosis evoked by that firing was also increased. However, exocytosis elicited by ACh did not vary between control and cirrhotic rats. Exocytosis triggered by depolarizing pulses was also unchanged. Amperometric recordings confirmed the lack of increased catecholamine charge released in cirrhosis after ACh or depolarization stimuli. However, the amperometric spikes exhibited faster kinetics of release. The overall Ca2+ entry through voltage-dependent Ca2+ channels (VDCC), or in particular through Cav1 channels, did not vary between chromaffin cells of control and cirrhotic rats. The inhibition of VDCC by methionine-enkephaline or ATP was not either altered, but it was increased by adrenaline in cells of cirrhotic rats. When a cocktail composed by the three neurotransmitters was tested in order to approach a situation closer to the physiological condition, the inhibition of VDCC was similar between both types of cells. In summary, chromaffin cells of the adrenal gland might contribute to exacerbate the sympathetic nervous system over-activity in cirrhosis because of an increased exocytosis elicited by an enhanced spontaneous electrical activity.


Assuntos
Potenciais de Ação/fisiologia , Células Cromafins/metabolismo , Exocitose/fisiologia , Cirrose Hepática/metabolismo , Animais , Canais de Cálcio/metabolismo , Tetracloreto de Carbono/toxicidade , Catecolaminas/metabolismo , Progressão da Doença , Cirrose Hepática/induzido quimicamente , Masculino , Ratos , Ratos Wistar
15.
Adv Exp Med Biol ; 1135: 105-117, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31098813

RESUMO

Cholesterol is the one of the major constituents of cell membranes providing these structures with a certain degree of rigidity. Proteins, such as ion channels, are molecules inserted in cell membranes and their activity is regulated by cholesterol and other molecules of a lipidic nature present in them. The molecular mechanisms underlying the regulation of ion channels by lipids and similar molecules have been an object of study for several years. A little over two decades ago, the first mammalian member of the Transient Receptor Potential (TRP) family of ion channels was cloned. This protein, the TRPV1 channel, was shown to integrate several types of noxious signals in sensory neurons and to participate in processes associated to the generation of pain. Thus, TRPV1 has become the target of intense research directed towards finding potential inhibitors of its activity in an effort to control pain. To date, several activators and positive modulators of the activity of TRPV1 have been described. However, very few naturally-occurring inhibitors are known. An endogenously-produced molecule that inhibits the activity of TRPV1 is cholesterol. This chapter focuses on describing the mechanisms by which the activity of TRPV1 can be regulated by this sterol.


Assuntos
Colesterol/química , Dor , Canais de Cátion TRPV/química , Animais , Lipídeos , Neurônios Aferentes
16.
J Neurophysiol ; 120(3): 1198-1211, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29947596

RESUMO

Lysophosphatidic acid (LPA) is a bioactive phospholipid that exhibits a wide array of functions that include regulation of protein synthesis and adequate development of organisms. LPA is present in the membranes of cells and in the serum of several mammals and has also been shown to participate importantly in pathophysiological conditions. For several decades it was known that LPA produces some of its effects in cells through its interaction with specific G protein-coupled receptors, which in turn are responsible for signaling pathways that regulate cellular function. Among the target proteins for LPA receptors are ion channels that modulate diverse aspects of the physiology of cells and organs where they are expressed. However, recent studies have begun to unveil direct effects of LPA on ion channels, highlighting this phospholipid as a direct agonist and adding to the knowledge of the field of lipid-protein interactions. Moreover, the roles of LPA in pathophysiological conditions associated with the function of some ion channels have also begun to be clarified, and molecular mechanisms have been identified. This review focuses on the effects of LPA on ion channel function under normal and pathological conditions and highlights our present knowledge of the mechanisms by which it regulates the function and expression of N- and T-type Ca++ channels; M-type K+ channel and inward rectifier K+ channel subunit 2.1; transient receptor potential (TRP) melastatin 2, TRP vanilloid 1, and TRP ankyrin 1 channels; and TWIK-related K+ channel 1 (TREK-1), TREK-2, TWIK-related spinal cord K+ channel (TRESK), and TWIK-related arachidonic acid-stimulated K+ channel (TRAAK).


Assuntos
Canais Iônicos/metabolismo , Lisofosfolipídeos/metabolismo , Dor/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Convulsões/metabolismo , Animais , Humanos , Lisofosfolipídeos/química , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
17.
Curr Top Membr ; 80: 139-161, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28863814

RESUMO

The transient receptor potential (TRP) family of ion channels is constituted by several nonselective cation channels that are activated by diverse stimuli and that function as polymodal receptors. TRP ion channels are expressed in neural and nonneural tissues where they play important roles in cell physiology. The activation of these ion channels is achieved through changes in temperature, osmolarity, voltage, pH, pressure, and by some natural or synthetic chemical compounds that directly bind to these proteins to regulate their activity. Other compounds that regulate TRP ion channel function are some endogenously synthetized lipid compounds. One such example of a compound of lipidic nature, commonly found in cells, is cholesterol. Cholesterol has been shown to exert positive and negative roles on TRP ion channel activity, albeit through different mechanisms which include a direct interaction of this molecule with specific amino acids located in the sequence of these channels or through the recruitment of the channels into specialized membrane microdomains (lipid rafts or caveolae). In this chapter, we will discuss important aspects of cholesterol as a regulator of some members of the TRP family of ion channels, and we will highlight the role of cholesterol on thermo-, chemo-, and osmosensation through regulation of the activity of these proteins.


Assuntos
Colesterol/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos , Canais de Potencial de Receptor Transitório/química
18.
J Biol Chem ; 289(35): 24079-90, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25035428

RESUMO

The transient receptor potential vanilloid 1 (TRPV1) ion channel is a polymodal protein that responds to various stimuli, including capsaicin (the pungent compound found in chili peppers), extracellular acid, and basic intracellular pH, temperatures close to 42 °C, and several lipids. Lysophosphatidic acid (LPA), an endogenous lipid widely associated with neuropathic pain, is an agonist of the TRPV1 channel found in primary afferent nociceptors and is activated by other noxious stimuli. Agonists or antagonists of lipid and other chemical natures are known to possess specific structural requirements for producing functional effects on their targets. To better understand how LPA and other lipid analogs might interact and affect the function of TRPV1, we set out to determine the structural features of these lipids that result in the activation of TRPV1. By changing the acyl chain length, saturation, and headgroup of these LPA analogs, we established strict requirements for activation of TRPV1. Among the natural LPA analogs, we found that only LPA 18:1, alkylglycerophosphate 18:1, and cyclic phosphatidic acid 18:1, all with a monounsaturated C18 hydrocarbon chain activate TRPV1, whereas polyunsaturated and saturated analogs do not. Thus, TRPV1 shows a more restricted ligand specificity compared with LPA G-protein-coupled receptors. We synthesized fatty alcohol phosphates and thiophosphates and found that many of them with a single double bond in position Δ9, 10, or 11 and Δ9 cyclopropyl group can activate TRPV1 with efficacy similar to capsaicin. Finally, we developed a pharmacophore and proposed a mechanistic model for how these lipids could induce a conformational change that activates TRPV1.


Assuntos
Lisofosfolipídeos/metabolismo , Canais de Cátion TRPV/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Conformação Proteica , Canais de Cátion TRPV/química
19.
Biochim Biophys Acta ; 1833(3): 698-711, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23220011

RESUMO

We recently characterized a nuclear import pathway for ß-dystroglycan; however, its nuclear role remains unknown. In this study, we demonstrate for the first time, the interaction of ß-dystroglycan with distinct proteins from different nuclear compartments, including the nuclear envelope (NE) (emerin and lamins A/C and B1), splicing speckles (SC35), Cajal bodies (p80-coilin), and nucleoli (Nopp140). Electron microscopy analysis revealed that ß-dystroglycan localized in the inner nuclear membrane, nucleoplasm, and nucleoli. Interestingly, downregulation of ß-dystroglycan resulted in both mislocalization and decreased expression of emerin and lamin B1, but not lamin A/C, as well in disorganization of nucleoli, Cajal bodies, and splicing speckles with the concomitant decrease in the levels of Nopp140, and p80-coilin, but not SC35. Quantitative reverse transcription PCR and cycloheximide-mediated protein arrest assays revealed that ß-dystroglycan deficiency did not change mRNA expression of NE proteins emerin and lamin B1 bud did alter their stability, accelerating protein turnover. Furthermore, knockdown of ß-dystroglycan disrupted NE-mediated processes including nuclear morphology and centrosome-nucleus linkage, which provides evidence that ß-dystroglycan association with NE proteins is biologically relevant. Unexpectedly, ß-dystroglycan-depleted cells exhibited multiple centrosomes, a characteristic of cancerous cells. Overall, these findings imply that ß-dystroglycan is a nuclear scaffolding protein involved in nuclear organization and NE structure and function, and that might be a contributor to the biogenesis of nuclear envelopathies.


Assuntos
Nucléolo Celular/metabolismo , Núcleo Celular/ultraestrutura , Corpos Enovelados/metabolismo , Distroglicanas/metabolismo , Mioblastos/metabolismo , Membrana Nuclear/metabolismo , Animais , Western Blotting , Nucléolo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Corpos Enovelados/genética , Distroglicanas/genética , Imunofluorescência , Imunoprecipitação , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mioblastos/citologia , Mioblastos/ultraestrutura , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Membr Biol ; 247(6): 479-91, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24676478

RESUMO

Transient receptor potential channels have been put forward as regulators of insulin secretion. A role for the TRPV1 ion channel in insulin secretion has been suggested in pancreatic beta cell lines. We explored whether TRPV1 is functionally expressed in RINm5F and primary beta cells from neonate and adult rats. We examined if capsaicin could activate cationic non-selective currents. Our results show that TRPV1 channels are not functional in insulin-secreting cells, since capsaicin did not produce current activation, not even under culture conditions known to induce the expression of other ion channels in these cells. Although TRPV1 channels seem to be irrelevant for the physiology of isolated beta cells, they may play a role in glucose homeostasis acting through the nerve fibers that regulate islet function. At the physiological level, we observed that Trpv1 (-/-) mice presented lower fasting insulin levels than their wild-type littermates, however, we did not find differences between these experimental groups nor in the glucose tolerance test or in the insulin secretion. However, we did find that the Trpv1 (-/-) mice exhibited a higher insulin sensitivity compared to their wild-type counterparts. Our results demonstrate that TRPV1 does not contribute to glucose-induced insulin secretion in beta cells as was previously thought, but it is possible that it may control insulin sensitivity.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Secreção de Insulina , Camundongos , Ratos , Canais de Cátion TRPV/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa