Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(15): e2322135121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38568964

RESUMO

Endothelial cells (ECs) line the wall of blood vessels and regulate arterial contractility to tune regional organ blood flow and systemic pressure. Chloride (Cl-) is the most abundant anion in ECs and the Cl- sensitive With-No-Lysine (WNK) kinase is expressed in this cell type. Whether intracellular Cl- signaling and WNK kinase regulate EC function to alter arterial contractility is unclear. Here, we tested the hypothesis that intracellular Cl- signaling in ECs regulates arterial contractility and examined the signaling mechanisms involved, including the participation of WNK kinase. Our data obtained using two-photon microscopy and cell-specific inducible knockout mice indicated that acetylcholine, a prototypical vasodilator, stimulated a rapid reduction in intracellular Cl- concentration ([Cl-]i) due to the activation of TMEM16A, a Cl- channel, in ECs of resistance-size arteries. TMEM16A channel-mediated Cl- signaling activated WNK kinase, which phosphorylated its substrate proteins SPAK and OSR1 in ECs. OSR1 potentiated transient receptor potential vanilloid 4 (TRPV4) currents in a kinase-dependent manner and required a conserved binding motif located in the channel C terminus. Intracellular Ca2+ signaling was measured in four dimensions in ECs using a high-speed lightsheet microscope. WNK kinase-dependent activation of TRPV4 channels increased local intracellular Ca2+ signaling in ECs and produced vasodilation. In summary, we show that TMEM16A channel activation reduces [Cl-]i, which activates WNK kinase in ECs. WNK kinase phosphorylates OSR1 which then stimulates TRPV4 channels to produce vasodilation. Thus, TMEM16A channels regulate intracellular Cl- signaling and WNK kinase activity in ECs to control arterial contractility.


Assuntos
Cloretos , Proteínas Serina-Treonina Quinases , Camundongos , Animais , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Cloretos/metabolismo , Células Endoteliais/metabolismo , Canais de Cátion TRPV/metabolismo , Transdução de Sinais/fisiologia
2.
Nat Commun ; 14(1): 1167, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36859399

RESUMO

Angelman syndrome (AS) is a neurogenetic disorder characterized by intellectual disability and atypical behaviors. AS results from loss of expression of the E3 ubiquitin-protein ligase UBE3A from the maternal allele in neurons. Individuals with AS display impaired coordination, poor balance, and gait ataxia. PIEZO2 is a mechanosensitive ion channel essential for coordination and balance. Here, we report that PIEZO2 activity is reduced in Ube3a deficient male and female mouse sensory neurons, a human Merkel cell carcinoma cell line and female human iPSC-derived sensory neurons with UBE3A knock-down, and de-identified stem cell-derived neurons from individuals with AS. We find that loss of UBE3A decreases actin filaments and reduces PIEZO2 expression and function. A linoleic acid (LA)-enriched diet increases PIEZO2 activity, mechano-excitability, and improves gait in male AS mice. Finally, LA supplementation increases PIEZO2 function in stem cell-derived neurons from individuals with AS. We propose a mechanism whereby loss of UBE3A expression reduces PIEZO2 function and identified a fatty acid that enhances channel activity and ameliorates AS-associated mechano-sensory deficits.


Assuntos
Síndrome de Angelman , Canais Iônicos , Ácido Linoleico , Animais , Feminino , Humanos , Masculino , Camundongos , Alelos , Síndrome de Angelman/tratamento farmacológico , Síndrome de Angelman/genética , Modelos Animais de Doenças , Deficiência Intelectual , Canais Iônicos/genética , Ácido Linoleico/farmacologia
3.
Cell Rep ; 40(10): 111306, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36070688

RESUMO

TRPV4 channel activation in endothelial cells leads to vasodilation, while impairment of TRPV4 activity is implicated in vascular dysfunction. Strategies that increase TRPV4 activity could enhance vasodilation and ameliorate vascular disorders. Here, we show that supplementation with eicosapentaenoic acid (EPA), an ω-3 polyunsaturated fatty acid known to have beneficial cardiovascular effects, increases TRPV4 activity in human endothelial cells of various vascular beds. Mice carrying the C. elegans FAT-1 enzyme, which converts ω-6 to ω-3 polyunsaturated fatty acids, display higher EPA content and increased TRPV4-mediated vasodilation in mesenteric arteries. Likewise, mice fed an EPA-enriched diet exhibit enhanced and prolonged TRPV4-dependent vasodilation in an endothelial cell-specific manner. We also show that EPA supplementation reduces TRPV4 desensitization, which contributes to the prolonged vasodilation. Neutralization of positive charges in the TRPV4 N terminus impairs the effect of EPA on channel desensitization. These findings highlight the beneficial effects of manipulating fatty acid content to enhance TRPV4-mediated vasodilation.


Assuntos
Ácidos Graxos Ômega-3 , Vasodilatação , Animais , Caenorhabditis elegans , Dieta , Células Endoteliais , Ácidos Graxos Ômega-3/farmacologia , Humanos , Camundongos , Canais de Cátion TRPV/genética
4.
Cell Rep ; 39(11): 110937, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35705057

RESUMO

Intestinal epithelial tight junction disruption is a primary contributing factor in alcohol-associated endotoxemia, systemic inflammation, and multiple organ damage. Ethanol and acetaldehyde disrupt tight junctions by elevating intracellular Ca2+. Here we identify TRPV6, a Ca2+-permeable channel, as responsible for alcohol-induced elevation of intracellular Ca2+, intestinal barrier dysfunction, and systemic inflammation. Ethanol and acetaldehyde elicit TRPV6 ionic currents in Caco-2 cells. Studies in Caco-2 cell monolayers and mouse intestinal organoids show that TRPV6 deficiency or inhibition attenuates ethanol- and acetaldehyde-induced Ca2+ influx, tight junction disruption, and barrier dysfunction. Moreover, Trpv6-/- mice are resistant to alcohol-induced intestinal barrier dysfunction. Photoaffinity labeling of 3-azibutanol identifies a histidine as a potential alcohol-binding site in TRPV6. The substitution of this histidine, and a nearby arginine, reduces ethanol-activated currents. Our findings reveal that TRPV6 is required for alcohol-induced gut barrier dysfunction and inflammation. Molecules that decrease TRPV6 function have the potential to attenuate alcohol-associated tissue injury.


Assuntos
Endotoxemia , Etanol , Histidina , Mucosa Intestinal , Canais de Cátion TRPV , Acetaldeído/toxicidade , Animais , Células CACO-2 , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Etanol/toxicidade , Histidina/farmacologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Camundongos , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo
5.
ACS Med Chem Lett ; 12(4): 572-578, 2021 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-33859797

RESUMO

The overactivation of transient receptor potential canonical 3 (TRPC3) is associated with neurodegenerative diseases and hypertension. Pyrazole 3 (Pyr3) is reported as the most selective TRPC3 inhibitor, but it has two inherent structural limitations: (1) the labile ester moiety leads to its rapid hydrolysis to the inactive Pyr8 in vivo, and (2) the alkylating trichloroacrylic amide moiety is known to be toxic. To circumvent these limitations, we designed a series of conformationally restricted Pyr3 analogues and reported that compound 20 maintains high potency and selectivity for human TRPC3 over its closely related TRP channels. It has significantly improved metabolic stability compared with Pyr3 and has a good safety profile. Preliminary evaluation of 20 demonstrated its ability to rescue Aß-induced neuron damage with similar potency to that of Pyr3 in vitro. Collectively, these results suggest that 20 represents a promising scaffold to potentially ameliorate the symptoms associated with TRPC3-mediated neurological and cardiovascular disorders.

6.
J Neurosci ; 41(3): 408-423, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33239401

RESUMO

Membrane remodeling by inflammatory mediators influences the function of sensory ion channels. The capsaicin- and heat-activated transient receptor potential vanilloid 1 (TRPV1) channel contributes to neurogenic inflammation and pain hypersensitivity, in part because of its potentiation downstream of phospholipase C-coupled receptors that regulate phosphoinositide lipid content. Here, we determined the effect of phosphoinositide lipids on TRPV1 function by combining genetic dissection, diet supplementation, and behavioral, biochemical, and functional analyses in Caenorhabditis elegans As capsaicin elicits heat and pain sensations in mammals, transgenic TRPV1 worms exhibit an aversive response to capsaicin. TRPV1 worms with low levels of phosphoinositide lipids display an enhanced response to capsaicin, whereas phosphoinositide lipid supplementation reduces TRPV1-mediated responses. A worm carrying a TRPV1 construct lacking the distal C-terminal domain features an enhanced response to capsaicin, independent of the phosphoinositide lipid content. Our results demonstrate that TRPV1 activity is enhanced when the phosphoinositide lipid content is reduced, and the C-terminal domain is key to determining agonist response in vivo.


Assuntos
Caenorhabditis elegans/fisiologia , Metabolismo dos Lipídeos , Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/deficiência , Canais de Cátion TRPV/fisiologia , Animais , Comportamento Animal , Proteínas de Caenorhabditis elegans/biossíntese , Sinalização do Cálcio/efeitos dos fármacos , Capsaicina/farmacologia , Dieta , Suplementos Nutricionais , Células HEK293 , Humanos , Neurônios/metabolismo , Fosfatidilinositóis/farmacologia , Canais de Cátion TRPV/genética
7.
Nat Commun ; 11(1): 3938, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753574

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Nat Commun ; 11(1): 2997, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32561714

RESUMO

PIEZO2 is the essential transduction channel for touch discrimination, vibration, and proprioception. Mice and humans lacking Piezo2 experience severe mechanosensory and proprioceptive deficits and fail to develop tactile allodynia. Bradykinin, a proalgesic agent released during inflammation, potentiates PIEZO2 activity. Molecules that decrease PIEZO2 function could reduce heightened touch responses during inflammation. Here, we find that the dietary fatty acid margaric acid (MA) decreases PIEZO2 function in a dose-dependent manner. Chimera analyses demonstrate that the PIEZO2 beam is a key region tuning MA-mediated channel inhibition. MA reduces neuronal action potential firing elicited by mechanical stimuli in mice and rat neurons and counteracts PIEZO2 sensitization by bradykinin. Finally, we demonstrate that this saturated fatty acid decreases PIEZO2 currents in touch neurons derived from human induced pluripotent stem cells. Our findings report on a natural product that inhibits PIEZO2 function and counteracts neuronal mechanical sensitization and reveal a key region for channel inhibition.


Assuntos
Ácidos Graxos/administração & dosagem , Canais Iônicos/antagonistas & inibidores , Mecanotransdução Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Propriocepção/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Algoritmos , Animais , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Canais Iônicos/genética , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Mecanotransdução Celular/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/fisiologia , Propriocepção/genética , Propriocepção/fisiologia , Ratos , Tato/efeitos dos fármacos , Tato/fisiologia
9.
J Cell Sci ; 132(23)2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31722978

RESUMO

TRP channels of the transient receptor potential ion channel superfamily are involved in a wide variety of mechanosensory processes, including touch sensation, pain, blood pressure regulation, bone loading and detection of cerebrospinal fluid flow. However, in many instances it is unclear whether TRP channels are the primary transducers of mechanical force in these processes. In this study, we tested stretch activation of eleven TRP channels from six mammalian subfamilies. We found that these TRP channels were insensitive to short membrane stretches in cellular systems. Furthermore, we purified TRPC6 and demonstrated its insensitivity to stretch in liposomes, an artificial bilayer system free from cellular components. Additionally, we demonstrated that, when expressed in C. elegans neurons, mouse TRPC6 restores the mechanoresponse of a touch insensitive mutant but requires diacylglycerol for activation. These results strongly suggest that the mammalian members of the TRP ion channel family are insensitive to tension induced by cell membrane stretching and, thus, are more likely to be activated by cytoplasmic tethers or downstream components and to act as amplifiers of cellular mechanosensory signaling cascades.


Assuntos
Canal de Cátion TRPC6/química , Animais , Células CHO , Caenorhabditis elegans/metabolismo , Cricetulus , Eletrofisiologia , Células HEK293 , Células HeLa , Humanos , Mecanotransdução Celular/fisiologia , Neurônios/metabolismo , Proteolipídeos/química
10.
Nat Commun ; 10(1): 1200, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30867417

RESUMO

Mechanosensitive ion channels rely on membrane composition to transduce physical stimuli into electrical signals. The Piezo1 channel mediates mechanoelectrical transduction and regulates crucial physiological processes, including vascular architecture and remodeling, cell migration, and erythrocyte volume. The identity of the membrane components that modulate Piezo1 function remain largely unknown. Using lipid profiling analyses, we here identify dietary fatty acids that tune Piezo1 mechanical response. We find that margaric acid, a saturated fatty acid present in dairy products and fish, inhibits Piezo1 activation and polyunsaturated fatty acids (PUFAs), present in fish oils, modulate channel inactivation. Force measurements reveal that margaric acid increases membrane bending stiffness, whereas PUFAs decrease it. We use fatty acid supplementation to abrogate the phenotype of gain-of-function Piezo1 mutations causing human dehydrated hereditary stomatocytosis. Beyond Piezo1, our findings demonstrate that cell-intrinsic lipid profile and changes in the fatty acid metabolism can dictate the cell's response to mechanical cues.


Assuntos
Anemia Hemolítica Congênita/dietoterapia , Gorduras na Dieta/metabolismo , Hidropisia Fetal/dietoterapia , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Anemia Hemolítica Congênita/genética , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/metabolismo , Ácidos Graxos Insaturados/administração & dosagem , Ácidos Graxos Insaturados/metabolismo , Mutação com Ganho de Função , Células HEK293 , Humanos , Hidropisia Fetal/genética , Canais Iônicos/genética , Metabolismo dos Lipídeos/fisiologia , Camundongos , Microscopia de Força Atômica , Técnicas de Patch-Clamp
11.
Stem Cell Rev Rep ; 15(1): 67-81, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30324358

RESUMO

The potential of human mesenchymal stromal/stem cells (MSCs) including oral stem cells (OSCs) as a cell source to derive functional neurons has been inconclusive. Here we tested a number of human OSCs for their neurogenic potential compared to non-OSCs and employed various neurogenic induction methods. OSCs including dental pulp stem cells (DPSCs), gingiva-derived mesenchymal stem cells (GMSCs), stem cells from apical papilla and non-OSCs including bone marrow MSCs (BMMSCs), foreskin fibroblasts and dermal fibroblasts using non-neurosphere-mediated or neurosphere-mediated methods to guide them toward neuronal lineages. Cells were subjected to RT-qPCR, immunocytofluorescence to detect the expression of neurogenic genes or electrophysiological analysis at final stage of maturation. We found that induced DPSCs and GMSCs overall appeared to be more neurogenic compared to other cells either morphologically or levels of neurogenic gene expression. Nonetheless, of all the neural induction methods employed, only one neurosphere-mediated method yielded electrophysiological properties of functional neurons. Under this method, cells expressed increased neural stem cell markers, nestin and SOX1, in the first phase of differentiation. Neuronal-like cells expressed ßIII-tubulin, CNPase, GFAP, MAP-2, NFM, pan-Nav, GAD67, Nav1.6, NF1, NSE, PSD95, and synapsin after the second phase of differentiation to maturity. Electrophysiological experiments revealed that 8.3% of DPSC-derived neuronal cells and 21.2% of GMSC-derived neuronal cells displayed action potential, although no spontaneous excitatory/inhibitory postsynaptic action potential was observed. We conclude that DPSCs and GMSCs have the potential to become neuronal cells in vitro, therefore, these cells may be used as a source for neural regeneration.


Assuntos
Potenciais de Ação/fisiologia , Diferenciação Celular , Polpa Dentária/citologia , Gengiva/citologia , Células-Tronco Mesenquimais/citologia , Neurogênese , Adolescente , Adulto , Células-Tronco Adultas/citologia , Diferenciação Celular/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Esferoides Celulares/citologia , Adulto Jovem
12.
J Biol Chem ; 293(41): 16102-16114, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30139744

RESUMO

The transient receptor potential ion channels support Ca2+ permeation in many organs, including the heart, brain, and kidney. Genetic mutations in transient receptor potential cation channel subfamily C member 3 (TRPC3) are associated with neurodegenerative diseases, memory loss, and hypertension. To better understand the conformational changes that regulate TRPC3 function, we solved the cryo-EM structures for the full-length human TRPC3 and its cytoplasmic domain (CPD) in the apo state at 5.8- and 4.0-Å resolution, respectively. These structures revealed that the TRPC3 transmembrane domain resembles those of other TRP channels and that the CPD is a stable module involved in channel assembly and gating. We observed the presence of a C-terminal domain swap at the center of the CPD where horizontal helices (HHs) transition into a coiled-coil bundle. Comparison of TRPC3 structures revealed that the HHs can reside in two distinct positions. Electrophysiological analyses disclosed that shortening the length of the C-terminal loop connecting the HH with the TRP helices increases TRPC3 activity and that elongating the length of the loop has the opposite effect. Our findings indicate that the C-terminal loop affects channel gating by altering the allosteric coupling between the cytoplasmic and transmembrane domains. We propose that molecules that target the HH may represent a promising strategy for controlling TRPC3-associated neurological disorders and hypertension.


Assuntos
Ativação do Canal Iônico , Canais de Cátion TRPC/química , Regulação Alostérica , Repetição de Anquirina , Células HEK293 , Humanos , Mutação , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Canais de Cátion TRPC/genética
13.
J Vis Exp ; (137)2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-30035769

RESUMO

Polymodal ion channels transduce multiple stimuli of different natures into allosteric changes; these dynamic conformations are challenging to determine and remain largely unknown. With recent advances in single-particle cryo-electron microscopy (cryo-EM) shedding light on the structural features of agonist binding sites and the activation mechanism of several ion channels, the stage is set for an in-depth dynamic analysis of their gating mechanisms using spectroscopic approaches. Spectroscopic techniques such as electron paramagnetic resonance (EPR) and double electron-electron resonance (DEER) have been mainly restricted to the study of prokaryotic ion channels that can be purified in large quantities. The requirement for large amounts of functional and stable membrane proteins has hampered the study of mammalian ion channels using these approaches. EPR and DEER offer many advantages, including determination of the structure and dynamic changes of mobile protein regions, albeit at low resolution, that might be difficult to obtain by X-ray crystallography or cryo-EM, and monitoring reversible gating transition (i.e., closed, open, sensitized, and desensitized). Here, we provide protocols for obtaining milligrams of functional detergent-solubilized transient receptor potential cation channel subfamily V member 1 (TRPV1) that can be labeled for EPR and DEER spectroscopy.


Assuntos
Análise Espectral/métodos , Canais de Cátion TRPV/genética , Células HEK293 , Humanos , Canais de Cátion TRPV/metabolismo
14.
J Biol Chem ; 293(26): 10381-10391, 2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-29752403

RESUMO

The kidney maintains the internal milieu by regulating the retention and excretion of proteins, ions, and small molecules. The glomerular podocyte forms the slit diaphragm of the ultrafiltration filter, whose damage leads to progressive kidney failure and focal segmental glomerulosclerosis (FSGS). The canonical transient receptor potential 6 (TRPC6) ion channel is expressed in the podocyte, and mutations in its cytoplasmic domain cause FSGS in humans. In vitro evaluation of disease-causing mutations in TRPC6 has revealed that these genetic alterations result in abnormal ion channel gating. However, the mechanism whereby the cytoplasmic domain modulates TRPC6 function is largely unknown. Here, we report a cryo-EM structure of the cytoplasmic domain of murine TRPC6 at 3.8 Å resolution. The cytoplasmic fold of TRPC6 is characterized by an inverted dome-like chamber pierced by four radial horizontal helices that converge into a vertical coiled-coil at the central axis. Unlike other TRP channels, TRPC6 displays a unique domain swap that occurs at the junction of the horizontal helices and coiled-coil. Multiple FSGS mutations converge at the buried interface between the vertical coiled-coil and the ankyrin repeats, which form the dome, suggesting these regions are critical for allosteric gating modulation. This functionally critical interface is a potential target for drug design. Importantly, dysfunction in other family members leads to learning deficits (TRPC1/4/5) and ataxia (TRPC3). Our data provide a structural framework for the mechanistic investigation of the TRPC family.


Assuntos
Microscopia Crioeletrônica , Citoplasma/metabolismo , Canal de Cátion TRPC6/química , Canal de Cátion TRPC6/metabolismo , Animais , Células HEK293 , Humanos , Camundongos , Mutação , Domínios Proteicos , Canal de Cátion TRPC6/genética
15.
Curr Opin Struct Biol ; 51: 92-98, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29602157

RESUMO

Membrane lipid composition and remodeling influence the function of ion channels. Polyunsaturated fatty acids (PUFAs) and their derivatives modulate ion channel function; whether this effect occurs directly by binding to the protein or indirectly through alteration of membranes' mechanical properties has been difficult to distinguish. There are a large number of studies addressing the effect of fatty acids; recent structural and functional analyses have identified binding sites and provided further evidence for the role of the plasma membrane in ion channel function. Here, we review cation channels that do not share a common topology or lipid-binding signature sequence, but for which there are recent compelling data that support both direct and indirect modulation by PUFAs or their derivatives.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Lipídeos de Membrana/metabolismo , Canais Iônicos/química , Lipídeos de Membrana/química , Ligação Proteica , Transdução de Sinais
16.
Cell Rep ; 21(1): 246-258, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28978477

RESUMO

Dietary consumption of ω-3 polyunsaturated fatty acids (PUFAs), present in fish oils, is known to improve the vascular response, but their molecular targets remain largely unknown. Activation of the TRPV4 channel has been implicated in endothelium-dependent vasorelaxation. Here, we studied the contribution of ω-3 PUFAs to TRPV4 function by precisely manipulating the fatty acid content in Caenorhabditis elegans. By genetically depriving the worms of PUFAs, we determined that the metabolism of ω-3 fatty acids is required for TRPV4 activity. Functional, lipid metabolome, and biophysical analyses demonstrated that ω-3 PUFAs enhance TRPV4 function in human endothelial cells and support the hypothesis that lipid metabolism and membrane remodeling regulate cell reactivity. We propose a model whereby the eicosanoid's epoxide group location increases membrane fluidity and influences the endothelial cell response by increasing TRPV4 channel activity. ω-3 PUFA-like molecules might be viable antihypertensive agents for targeting TRPV4 to reduce systemic blood pressure.


Assuntos
Anti-Hipertensivos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Canais de Cátion TRPV/genética , Animais , Animais Geneticamente Modificados , Anti-Hipertensivos/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Linhagem Celular , Membrana Celular/química , Membrana Celular/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Expressão Gênica , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fluidez de Membrana/efeitos dos fármacos , Metaboloma , Forbóis/farmacologia , Fosfolipídeos/metabolismo , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/metabolismo
17.
Sci Rep ; 7(1): 9861, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852163

RESUMO

The transient receptor potential vanilloid 1 (TRPV1) channel is an essential component of the cellular mechanism through which noxious stimuli evoke pain. Functional and structural characterizations of TRPV1 shed light on vanilloid activation, yet the mechanisms for temperature and proton gating remain largely unknown. Spectroscopic approaches are needed to understand the mechanisms by which TRPV1 translates diverse stimuli into channel opening. Here, we have engineered a minimal cysteine-less rat TRPV1 construct (eTRPV1) that can be stably purified and reconstituted for spectroscopic studies. Biophysical analyses of TRPV1 constructs reveal that the S5-pore helix loop influences protein stability and vanilloid and proton responses, but not thermal sensitivity. Cysteine mutants retain function and stability for double electron-electron resonance (DEER) and electron paramagnetic resonance (EPR) spectroscopies. DEER measurements in the closed state demonstrate that eTRPV1 reports distances in the extracellular vestibule, equivalent to those observed in the apo TRPV1 structure. EPR measurements show a distinct pattern of mobilities and spectral features, in detergent and liposomes, for residues at the pore domain that agree with their location in the TRPV1 structure. Our results set the stage for a systematic characterization of TRPV1 using spectroscopic approaches to reveal conformational changes compatible with thermal- and ligand-dependent gating.


Assuntos
Expressão Gênica , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/isolamento & purificação , Sequência de Aminoácidos , Animais , Cisteína/química , Simulação de Dinâmica Molecular , Mutação , Fosforilação , Conformação Proteica , Estabilidade Proteica , Prótons , Ratos , Proteínas Recombinantes , Análise Espectral , Canais de Cátion TRPV/química , Xenopus
18.
Neuron ; 77(4): 667-79, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23439120

RESUMO

The capsaicin receptor, TRPV1, is regulated by phosphatidylinositol-4,5-bisphosphate (PIP(2)), although the precise nature of this effect (i.e., positive or negative) remains controversial. Here, we reconstitute purified TRPV1 into artificial liposomes, where it is gated robustly by capsaicin, protons, spider toxins, and, notably, heat, demonstrating intrinsic sensitivity of the channel to both chemical and thermal stimuli. TRPV1 is fully functional in the absence of phosphoinositides, arguing against their proposed obligatory role in channel activation. Rather, introduction of various phosphoinositides, including PIP(2), PI4P, and phosphatidylinositol, inhibits TRPV1, supporting a model whereby phosphoinositide turnover contributes to thermal hyperalgesia by disinhibiting the channel. Using an orthogonal chemical strategy, we show that association of the TRPV1 C terminus with the bilayer modulates channel gating, consistent with phylogenetic data implicating this domain as a key regulatory site for tuning stimulus sensitivity. Beyond TRPV1, these findings are relevant to understanding how membrane lipids modulate other "receptor-operated" TRP channels.


Assuntos
Temperatura Alta/efeitos adversos , Canais Iônicos/metabolismo , Lipídeos/fisiologia , Fosfatidilinositóis/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/farmacologia , Células Cultivadas , Células Sf9/metabolismo , Spodoptera
19.
Structure ; 20(8): 1332-42, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22771214

RESUMO

In K+ channels, rearrangements of the pore outer vestibule have been associated with C-type inactivation gating. Paradoxically, the crystal structure of Open/C-type inactivated KcsA suggests these movements to be modest in magnitude. In this study, we show that under physiological conditions, the KcsA outer vestibule undergoes relatively large dynamic rearrangements upon inactivation. External Cd2+ enhances the rate of C-type inactivation in an cysteine mutant (Y82C) via metal-bridge formation. This effect is not present in a non-inactivating mutant (E71A/Y82C). Tandem dimer and tandem tetramer constructs of equivalent cysteine mutants in KcsA and Shaker K+ channels demonstrate that these Cd2+ metal bridges are formed only between adjacent subunits. This is well supported by molecular dynamics simulations. Based on the crystal structure of Cd2+ -bound Y82C-KcsA in the closed state, together with electron paramagnetic resonance distance measurements in the KcsA outer vestibule, we suggest that subunits must dynamically come in close proximity as the channels undergo inactivation.


Assuntos
Proteínas de Bactérias/química , Cádmio/química , Complexos de Coordenação/química , Canais de Potássio/química , Motivos de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Lipossomos/química , Simulação de Dinâmica Molecular , Canais de Potássio/genética , Estabilidade Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Streptomyces lividans , Termodinâmica
20.
J Biol Chem ; 286(45): 39091-9, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21908602

RESUMO

Emerging evidence suggests that K(+) channel inactivation involves coupling between residues in adjacent regions of the channel. Human ether-a-go-go-related gene-1 (hERG1) K(+) channels undergo a fast inactivation gating process that is crucial for maintaining electrical stability in the heart. The molecular mechanisms that drive inactivation in hERG1 channels are unknown. Using alanine scanning mutagenesis, we show that a pore helix residue (Thr-618) that points toward the S5 segment is critical for normal inactivation gating. Amino acid substitutions at position 618 modulate the free energy of inactivation gating, causing enhanced or reduced inactivation. Mutation of an S5 residue that is predicted to be adjacent to Thr-618 (W568L) abolishes inactivation and alters ion selectivity. The introduction of the Thr-618-equivalent residue in Kv1.5 enhances inactivation. Molecular dynamic simulations of the Kv1.2 tetramer reveal van der Waals coupling between hERG1 618- and 568-equivalent residues and a significant increase in interaction energies when threonine is introduced at the 618-equivalent position. We propose that coupling between the S5 segment and pore helix may participate in the inactivation process in hERG1 channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Ativação do Canal Iônico/fisiologia , Proteínas Musculares/metabolismo , Substituição de Aminoácidos , Animais , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Modelos Moleculares , Proteínas Musculares/genética , Mutagênese , Mutação de Sentido Incorreto , Miocárdio/metabolismo , Estrutura Secundária de Proteína , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...