Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(30): e2221958120, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37459546

RESUMO

Osteoarthritis is a chronic disease that can be initiated by altered joint loading or injury of the cartilage. The mechanically sensitive PIEZO ion channels have been shown to transduce injurious levels of biomechanical strain in articular chondrocytes and mediate cell death. However, the mechanisms of channel gating in response to high cellular deformation and the strain thresholds for activating PIEZO channels remain unclear. We coupled studies of single-cell compression using atomic force microscopy (AFM) with finite element modeling (FEM) to identify the biophysical mechanisms of PIEZO-mediated calcium (Ca2+) signaling in chondrocytes. We showed that PIEZO1 and PIEZO2 are needed for initiating Ca2+ signaling at moderately high levels of cellular deformation, but at the highest strains, PIEZO1 functions independently of PIEZO2. Biophysical factors that increase apparent chondrocyte membrane tension, including hypoosmotic prestrain, high compression magnitudes, and low deformation rates, also increased PIEZO1-driven Ca2+ signaling. Combined AFM/FEM studies showed that 50% of chondrocytes exhibit Ca2+ signaling at 80 to 85% nominal cell compression, corresponding to a threshold of apparent membrane finite principal strain of E = 1.31, which represents a membrane stretch ratio (λ) of 1.9. Both intracellular and extracellular Ca2+ are necessary for the PIEZO1-mediated Ca2+ signaling response to compression. Our results suggest that PIEZO1-induced signaling drives chondrocyte mechanical injury due to high membrane tension, and this threshold can be altered by factors that influence membrane prestress, such as cartilage hypoosmolarity, secondary to proteoglycan loss. These findings suggest that modulating PIEZO1 activation or downstream signaling may offer avenues for the prevention or treatment of osteoarthritis.


Assuntos
Condrócitos , Osteoartrite , Humanos , Condrócitos/metabolismo , Canais Iônicos/metabolismo , Articulações , Osteoartrite/metabolismo , Mecanotransdução Celular , Sinalização do Cálcio
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33758095

RESUMO

Osteoarthritis (OA) is a painful and debilitating condition of synovial joints without any disease-modifying therapies [A. M. Valdes, T. D. Spector, Nat. Rev. Rheumatol. 7, 23-32 (2011)]. We previously identified mechanosensitive PIEZO channels, PIEZO1 and PIEZO2, both expressed in articular cartilage, to function in chondrocyte mechanotransduction in response to injury [W. Lee et al., Proc. Natl. Acad. Sci. U.S.A. 111, E5114-E5122 (2014); W. Lee, F. Guilak, W. Liedtke, Curr. Top. Membr. 79, 263-273 (2017)]. We therefore asked whether interleukin-1-mediated inflammatory signaling, as occurs in OA, influences Piezo gene expression and channel function, thus indicative of maladaptive reprogramming that can be rationally targeted. Primary porcine chondrocyte culture and human osteoarthritic cartilage tissue were studied. We found that interleukin-1α (IL-1α) up-regulated Piezo1 in porcine chondrocytes. Piezo1 expression was significantly increased in human osteoarthritic cartilage. Increased Piezo1 expression in chondrocytes resulted in a feed-forward pathomechanism whereby increased function of Piezo1 induced excess intracellular Ca2+ at baseline and in response to mechanical deformation. Elevated resting state Ca2+ in turn rarefied the F-actin cytoskeleton and amplified mechanically induced deformation microtrauma. As intracellular substrates of this OA-related inflammatory pathomechanism, in porcine articular chondrocytes exposed to IL-1α, we discovered that enhanced Piezo1 expression depended on p38 MAP-kinase and transcription factors HNF4 and ATF2/CREBP1. CREBP1 directly bound to the proximal PIEZO1 gene promoter. Taken together, these signaling and genetic reprogramming events represent a detrimental Ca2+-driven feed-forward mechanism that can be rationally targeted to stem the progression of OA.


Assuntos
Condrócitos/metabolismo , Interleucina-1alfa/metabolismo , Canais Iônicos/genética , Mecanotransdução Celular/imunologia , Osteoartrite/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Cálcio/metabolismo , Cartilagem Articular/citologia , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/imunologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Osteoartrite/genética , Osteoartrite/patologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Sus scrofa , Regulação para Cima/imunologia
3.
J Physiol ; 601(9): 1655-1673, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36625071

RESUMO

The Transient Receptor Potential Vanilloid 4 (TRPV4) channel has been shown to function in many physiological and pathophysiological processes. Despite abundant information on its importance in physiology, very few endogenous agonists for this channel have been described, and very few underlying mechanisms for its activation have been clarified. TRPV4 is expressed by several types of cells, such as vascular endothelial, and skin and lung epithelial cells, where it plays pivotal roles in their function. In the present study, we show that TRPV4 is activated by lysophosphatidic acid (LPA) in both endogenous and heterologous expression systems, pinpointing this molecule as one of the few known endogenous agonists for TRPV4. Importantly, LPA is a bioactive glycerophospholipid, relevant in several physiological conditions, including inflammation and vascular function, where TRPV4 has also been found to be essential. Here we also provide mechanistic details of the activation of TRPV4 by LPA and another glycerophospholipid, lysophosphatidylcholine (LPC), and show that LPA directly interacts with both the N- and C-terminal regions of TRPV4 to activate this channel. Moreover, we show that LPC activates TRPV4 by producing an open state with a different single-channel conductance to that observed with LPA. Our data suggest that the activation of TRPV4 can be finely tuned in response to different endogenous lipids, highlighting this phenomenon as a regulator of cell and organismal physiology. KEY POINTS: The Transient Receptor Potential Vaniloid (TRPV) 4 ion channel is a widely distributed protein with important roles in normal and disease physiology for which few endogenous ligands are known. TRPV4 is activated by a bioactive lipid, lysophosphatidic acid (LPA) 18:1, in a dose-dependent manner, in both a primary and a heterologous expression system. Activation of TRPV4 by LPA18:1 requires residues in the N- and C-termini of the ion channel. Single-channel recordings show that TRPV4 is activated with a decreased current amplitude (conductance) in the presence of lysophosphatidylcholine (LPC) 18:1, while LPA18:1 and GSK101 activate the channel with a larger single-channel amplitude. Distinct single-channel amplitudes produced by LPA18:1 and LPC18:1 could differentially modulate the responses of the cells expressing TRPV4 under different physiological conditions.


Assuntos
Canais de Potencial de Receptor Transitório , Canais de Cátion TRPV/metabolismo , Lisofosfatidilcolinas/farmacologia , Lisofosfolipídeos/farmacologia
4.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1239-L1243, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401673

RESUMO

Lethality of coronavirus disease (COVID-19) during the 2020 pandemic, currently still in the exponentially accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We argue for inhibition of the transient receptor potential vanilloid 4 (TRPV4) calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in COVID-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in COVID-19 patients with respiratory malfunction and at risk for lung edema. Perplexingly, among the currently pursued therapeutic strategies against COVID-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce COVID-19 lethality but will also preempt a distressing healthcare scenario with insufficient capacity to provide ventilator-assisted respiration.


Assuntos
Betacoronavirus , Infecções por Coronavirus , Pulmão/virologia , Pandemias , Pneumonia Viral , Edema Pulmonar/prevenção & controle , Canais de Cátion TRPV/antagonistas & inibidores , COVID-19 , Cálcio/metabolismo , Infecções por Coronavirus/virologia , Humanos , Pulmão/metabolismo , Pneumonia Viral/virologia , Edema Pulmonar/virologia , Respiração Artificial , SARS-CoV-2
5.
J Biol Chem ; 291(19): 10252-62, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26961876

RESUMO

TRPV4 ion channels function in epidermal keratinocytes and in innervating sensory neurons; however, the contribution of the channel in either cell to neurosensory function remains to be elucidated. We recently reported TRPV4 as a critical component of the keratinocyte machinery that responds to ultraviolet B (UVB) and functions critically to convert the keratinocyte into a pain-generator cell after excess UVB exposure. One key mechanism in keratinocytes was increased expression and secretion of endothelin-1, which is also a known pruritogen. Here we address the question of whether TRPV4 in skin keratinocytes functions in itch, as a particular form of "forefront" signaling in non-neural cells. Our results support this novel concept based on attenuated scratching behavior in response to histaminergic (histamine, compound 48/80, endothelin-1), not non-histaminergic (chloroquine) pruritogens in Trpv4 keratinocyte-specific and inducible knock-out mice. We demonstrate that keratinocytes rely on TRPV4 for calcium influx in response to histaminergic pruritogens. TRPV4 activation in keratinocytes evokes phosphorylation of mitogen-activated protein kinase, ERK, for histaminergic pruritogens. This finding is relevant because we observed robust anti-pruritic effects with topical applications of selective inhibitors for TRPV4 and also for MEK, the kinase upstream of ERK, suggesting that calcium influx via TRPV4 in keratinocytes leads to ERK-phosphorylation, which in turn rapidly converts the keratinocyte into an organismal itch-generator cell. In support of this concept we found that scratching behavior, evoked by direct intradermal activation of TRPV4, was critically dependent on TRPV4 expression in keratinocytes. Thus, TRPV4 functions as a pruriceptor-TRP in skin keratinocytes in histaminergic itch, a novel basic concept with translational-medical relevance.


Assuntos
Sinalização do Cálcio , Epiderme/metabolismo , Histamina/metabolismo , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases , Prurido/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Endotelina-1/biossíntese , Endotelina-1/genética , Epiderme/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Histamina/genética , Queratinócitos/patologia , Camundongos , Camundongos Knockout , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Especificidade de Órgãos/efeitos da radiação , Prurido/tratamento farmacológico , Prurido/genética , Prurido/patologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética , Raios Ultravioleta/efeitos adversos
6.
Proc Natl Acad Sci U S A ; 111(4): 1316-21, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24474754

RESUMO

Mechanical loading of joints plays a critical role in maintaining the health and function of articular cartilage. The mechanism(s) of chondrocyte mechanotransduction are not fully understood, but could provide important insights into new physical or pharmacologic therapies for joint diseases. Transient receptor potential vanilloid 4 (TRPV4), a Ca(2+)-permeable osmomechano-TRP channel, is highly expressed in articular chondrocytes, and loss of TRPV4 function is associated with joint arthropathy and osteoarthritis. The goal of this study was to examine the hypothesis that TRPV4 transduces dynamic compressive loading in articular chondrocytes. We first confirmed the presence of physically induced, TRPV4-dependent intracellular Ca(2+) signaling in agarose-embedded chondrocytes, and then used this model system to study the role of TRPV4 in regulating the response of chondrocytes to dynamic compression. Inhibition of TRPV4 during dynamic loading prevented acute, mechanically mediated regulation of proanabolic and anticatabolic genes, and furthermore, blocked the loading-induced enhancement of matrix accumulation and mechanical properties. Furthermore, chemical activation of TRPV4 by the agonist GSK1016790A in the absence of mechanical loading similarly enhanced anabolic and suppressed catabolic gene expression, and potently increased matrix biosynthesis and construct mechanical properties. These findings support the hypothesis that TRPV4-mediated Ca(2+) signaling plays a central role in the transduction of mechanical signals to support cartilage extracellular matrix maintenance and joint health. Moreover, these insights raise the possibility of therapeutically targeting TRPV4-mediated mechanotransduction for the treatment of diseases such as osteoarthritis, as well as to enhance matrix formation and functional properties of tissue-engineered cartilage as an alternative to bioreactor-based mechanical stimulation.


Assuntos
Condrócitos/metabolismo , Mecanotransdução Celular/fisiologia , Canais de Cátion TRPV/fisiologia , Animais , Células Cultivadas , Condrócitos/citologia , Regulação da Expressão Gênica , Sefarose , Suínos
7.
Proc Natl Acad Sci U S A ; 111(47): E5114-22, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385580

RESUMO

Diarthrodial joints are essential for load bearing and locomotion. Physiologically, articular cartilage sustains millions of cycles of mechanical loading. Chondrocytes, the cells in cartilage, regulate their metabolic activities in response to mechanical loading. Pathological mechanical stress can lead to maladaptive cellular responses and subsequent cartilage degeneration. We sought to deconstruct chondrocyte mechanotransduction by identifying mechanosensitive ion channels functioning at injurious levels of strain. We detected robust expression of the recently identified mechanosensitive channels, PIEZO1 and PIEZO2. Combined directed expression of Piezo1 and -2 sustained potentiated mechanically induced Ca(2+) signals and electrical currents compared with single-Piezo expression. In primary articular chondrocytes, mechanically evoked Ca(2+) transients produced by atomic force microscopy were inhibited by GsMTx4, a PIEZO-blocking peptide, and by Piezo1- or Piezo2-specific siRNA. We complemented the cellular approach with an explant-cartilage injury model. GsMTx4 reduced chondrocyte death after mechanical injury, suggesting a possible therapy for reducing cartilage injury and posttraumatic osteoarthritis by attenuating Piezo-mediated cartilage mechanotransduction of injurious strains.


Assuntos
Cartilagem Articular/fisiologia , Canais Iônicos/fisiologia , Estresse Mecânico , Animais , Sinalização do Cálcio , Condrócitos/fisiologia , Canais Iônicos/genética , Camundongos , RNA Interferente Pequeno
8.
Am J Respir Cell Mol Biol ; 54(3): 370-83, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26222277

RESUMO

The cation channel transient receptor potential vanilloid (TRPV) 4 is expressed in endothelial and immune cells; however, its role in acute lung injury (ALI) is unclear. The functional relevance of TRPV4 was assessed in vivo, in isolated murine lungs, and in isolated neutrophils. Genetic deficiency of TRPV4 attenuated the functional, histological, and inflammatory hallmarks of acid-induced ALI. Similar protection was obtained with prophylactic administration of the TRPV4 inhibitor, GSK2193874; however, therapeutic administration of the TRPV4 inhibitor, HC-067047, after ALI induction had no beneficial effect. In isolated lungs, platelet-activating factor (PAF) increased vascular permeability in lungs perfused with trpv4(+/+) more than with trpv4(-/-) blood, independent of lung genotype, suggesting a contribution of TRPV4 on blood cells to lung vascular barrier failure. In neutrophils, TRPV4 inhibition or deficiency attenuated the PAF-induced increase in intracellular calcium. PAF induced formation of epoxyeicosatrienoic acids by neutrophils, which, in turn, stimulated TRPV4-dependent Ca(2+) signaling, whereas inhibition of epoxyeicosatrienoic acid formation inhibited the Ca(2+) response to PAF. TRPV4 deficiency prevented neutrophil responses to proinflammatory stimuli, including the formation of reactive oxygen species, neutrophil adhesion, and chemotaxis, putatively due to reduced activation of Rac. In chimeric mice, however, the majority of protective effects in acid-induced ALI were attributable to genetic deficiency of TRPV4 in parenchymal tissue, whereas TRPV4 deficiency in circulating blood cells primarily reduced lung myeloperoxidase activity. Our findings identify TRPV4 as novel regulator of neutrophil activation and suggest contributions of both parenchymal and neutrophilic TRPV4 in the pathophysiology of ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Pulmão/metabolismo , Ativação de Neutrófilo , Neutrófilos/metabolismo , Canais de Cátion TRPV/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/prevenção & controle , Animais , Transplante de Medula Óssea , Sinalização do Cálcio , Permeabilidade Capilar , Modelos Animais de Doenças , Humanos , Ácido Clorídrico , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Masculino , Camundongos Knockout , Morfolinas/farmacologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Pneumonia/metabolismo , Edema Pulmonar/metabolismo , Pirróis/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética
9.
Proc Natl Acad Sci U S A ; 110(15): 6157-62, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23530219

RESUMO

In the CNS, astrocytes are sensory and regulatory hubs that play important roles in cerebral homeostatic processes, including matching local cerebral blood flow to neuronal metabolism (neurovascular coupling). These cells possess a highly branched network of processes that project from the soma to neuronal synapses as well as to arterioles and capillaries, where they terminate in "endfeet" that encase the blood vessels. Ca(2+) signaling within the endfoot mediates neurovascular coupling; thus, these functional microdomains control vascular tone and local perfusion in the brain. Transient receptor potential vanilloid 4 (TRPV4) channels--nonselective cation channels with considerable Ca(2+) conductance--have been identified in astrocytes, but their function is largely unknown. We sought to characterize the influence of TRPV4 channels on Ca(2+) dynamics in the astrocytic endfoot microdomain and assess their role in neurovascular coupling. We identified local TRPV4-mediated Ca(2+) oscillations in endfeet and further found that TRPV4 Ca(2+) signals are amplified and propagated by Ca(2+)-induced Ca(2+) release from inositol trisphosphate receptors (IP3Rs). Moreover, TRPV4-mediated Ca(2+) influx contributes to the endfoot Ca(2+) response to neuronal activation, enhancing the accompanying vasodilation. Our results identify a dynamic synergy between TRPV4 channels and IP3Rs in astrocyte endfeet and demonstrate that TRPV4 channels are engaged in and contribute to neurovascular coupling.


Assuntos
Astrócitos/citologia , Cálcio/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Sinalização do Cálcio , Sistema Nervoso Central/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Oscilometria
10.
Proc Natl Acad Sci U S A ; 110(11): 4315-20, 2013 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-23440186

RESUMO

Bisphenol A (BPA) is a ubiquitous compound that is emerging as a possible toxicant during embryonic development. BPA has been shown to epigenetically affect the developing nervous system, but the molecular mechanisms are not clear. Here we demonstrate that BPA exposure in culture led to delay in the perinatal chloride shift caused by significant decrease in potassium chloride cotransporter 2 (Kcc2) mRNA expression in developing rat, mouse, and human cortical neurons. Neuronal chloride increased correspondingly. Treatment with epigenetic compounds decitabine and trichostatin A rescued the BPA effects as did knockdown of histone deacetylase 1 and combined knockdown histone deacetylase 1 and 2. Furthermore, BPA evoked increase in tangential interneuron migration and increased chloride in migrating neurons. Interestingly, BPA exerted its effect in a sexually dimorphic manner, with a more accentuated effect in females than males. By chromatin immunoprecipitation, we found a significant increase in binding of methyl-CpG binding protein 2 to the "cytosine-phosphate-guanine shores" of the Kcc2 promoter, and decrease in binding of acetylated histone H3K9 surrounding the transcriptional start site. Methyl-CpG binding protein 2-expressing neurons were more abundant resulting from BPA exposure. The sexually dimorphic effect of BPA on Kcc2 expression was also demonstrated in cortical neurons cultured from the offspring of BPA-fed mouse dams. In these neurons and in cortical slices, decitabine was found to rescue the effect of BPA on Kcc2 expression. Overall, our results indicate that BPA can disrupt Kcc2 gene expression through epigenetic mechanisms. Beyond increase in basic understanding, our findings have relevance for identifying unique neurodevelopmental toxicity mechanisms of BPA, which could possibly play a role in pathogenesis of human neurodevelopmental disorders.


Assuntos
Poluentes Ocupacionais do Ar/efeitos adversos , Compostos Benzidrílicos/efeitos adversos , Córtex Cerebral/metabolismo , Cloretos/metabolismo , Epigênese Genética/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Fenóis/efeitos adversos , Elementos de Resposta , Simportadores/biossíntese , Poluentes Ocupacionais do Ar/farmacologia , Animais , Compostos Benzidrílicos/farmacologia , Células Cultivadas , Doenças do Sistema Nervoso Central/induzido quimicamente , Doenças do Sistema Nervoso Central/metabolismo , Córtex Cerebral/patologia , Proteínas de Ligação a DNA/metabolismo , Feminino , Histona Desacetilase 1/metabolismo , Humanos , Masculino , Camundongos , Neurônios/patologia , Fenóis/farmacologia , Ratos , Caracteres Sexuais , Cotransportadores de K e Cl-
11.
Proc Natl Acad Sci U S A ; 110(34): E3225-34, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23929777

RESUMO

At our body surface, the epidermis absorbs UV radiation. UV overexposure leads to sunburn with tissue injury and pain. To understand how, we focus on TRPV4, a nonselective cation channel highly expressed in epithelial skin cells and known to function in sensory transduction, a property shared with other transient receptor potential channels. We show that following UVB exposure mice with induced Trpv4 deletions, specifically in keratinocytes, are less sensitive to noxious thermal and mechanical stimuli than control animals. Exploring the mechanism, we find that epidermal TRPV4 orchestrates UVB-evoked skin tissue damage and increased expression of the proalgesic/algogenic mediator endothelin-1. In culture, UVB causes a direct, TRPV4-dependent Ca(2+) response in keratinocytes. In mice, topical treatment with a TRPV4-selective inhibitor decreases UVB-evoked pain behavior, epidermal tissue damage, and endothelin-1 expression. In humans, sunburn enhances epidermal expression of TRPV4 and endothelin-1, underscoring the potential of keratinocyte-derived TRPV4 as a therapeutic target for UVB-induced sunburn, in particular pain.


Assuntos
Endotelina-1/metabolismo , Células Epiteliais/efeitos da radiação , Dor/metabolismo , Transdução de Sinais/efeitos da radiação , Queimadura Solar/metabolismo , Canais de Cátion TRPV/metabolismo , Raios Ultravioleta , Análise de Variância , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Dor/etiologia , Pele/citologia , Queimadura Solar/patologia
12.
Proc Natl Acad Sci U S A ; 114(8): 1765-1767, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28179562
13.
Proc Natl Acad Sci U S A ; 108(30): 12509-14, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21746918

RESUMO

Sodium appetite is an instinct that involves avid specific intention. It is elicited by sodium deficiency, stress-evoked adrenocorticotropic hormone (ACTH), and reproduction. Genome-wide microarrays in sodium-deficient mice or after ACTH infusion showed up-regulation of hypothalamic genes, including dopamine- and cAMP-regulated neuronal phosphoprotein 32 kDa (DARPP-32), dopamine receptors-1 and -2, α-2C- adrenoceptor, and striatally enriched protein tyrosine phosphatase (STEP). Both DARPP-32 and neural plasticity regulator activity-regulated cytoskeleton associated protein (ARC) were up-regulated in lateral hypothalamic orexinergic neurons by sodium deficiency. Administration of dopamine D1 (SCH23390) and D2 receptor (raclopride) antagonists reduced gratification of sodium appetite triggered by sodium deficiency. SCH23390 was specific, having no effect on osmotic-induced water drinking, whereas raclopride also reduced water intake. D1 receptor KO mice had normal sodium appetite, indicating compensatory regulation. Appetite was insensitive to SCH23390, confirming the absence of off-target effects. Bilateral microinjection of SCH23390 (100 nM in 200 nL) into rats' lateral hypothalamus greatly reduced sodium appetite. Gene set enrichment analysis in hypothalami of mice with sodium appetite showed significant enrichment of gene sets previously linked to addiction (opiates and cocaine). This finding of concerted gene regulation was attenuated on gratification with perplexingly rapid kinetics of only 10 min, anteceding significant absorption of salt from the gut. Salt appetite and hedonic liking of salt taste have evolved over >100 million y (e.g., being present in Metatheria). Drugs causing pleasure and addiction are comparatively recent and likely reflect usurping of evolutionary ancient systems with high survival value by the gratification of contemporary hedonic indulgences. Our findings outline a molecular logic for instinctive behavior encoded by the brain with possible important translational-medical implications.


Assuntos
Apetite/genética , Comportamento Aditivo/genética , Hipotálamo/fisiologia , Sódio na Dieta/administração & dosagem , Hormônio Adrenocorticotrópico/administração & dosagem , Hormônio Adrenocorticotrópico/fisiologia , Animais , Apetite/efeitos dos fármacos , Apetite/fisiologia , Comportamento Aditivo/fisiopatologia , Evolução Biológica , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/genética , Ingestão de Líquidos/fisiologia , Feminino , Estudo de Associação Genômica Ampla , Hipotálamo/efeitos dos fármacos , Instinto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Psicológicos , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Recompensa
14.
Sci Adv ; 7(5)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33571125

RESUMO

Mechanobiologic signals regulate cellular responses under physiologic and pathologic conditions. Using synthetic biology and tissue engineering, we developed a mechanically responsive bioartificial tissue that responds to mechanical loading to produce a preprogrammed therapeutic biologic drug. By deconstructing the signaling networks induced by activation of the mechanically sensitive ion channel transient receptor potential vanilloid 4 (TRPV4), we created synthetic TRPV4-responsive genetic circuits in chondrocytes. We engineered these cells into living tissues that respond to mechanical loading by producing the anti-inflammatory biologic drug interleukin-1 receptor antagonist. Chondrocyte TRPV4 is activated by osmotic loading and not by direct cellular deformation, suggesting that tissue loading is transduced into an osmotic signal that activates TRPV4. Either osmotic or mechanical loading of tissues transduced with TRPV4-responsive circuits protected constructs from inflammatory degradation by interleukin-1α. This synthetic mechanobiology approach was used to develop a mechanogenetic system to enable long-term, autonomously regulated drug delivery driven by physiologically relevant loading.


Assuntos
Produtos Biológicos , Canais de Cátion TRPV , Produtos Biológicos/metabolismo , Condrócitos/metabolismo , Redes Reguladoras de Genes , Canais de Cátion TRPV/metabolismo , Engenharia Tecidual
15.
SSRN ; : 3558887, 2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32714108

RESUMO

Lethality of Covid-19 during the 2020 pandemic, currently in the exponentially-accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of SARS-CoV-2 infection. We argue for inhibition of the TRPV4 calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema, and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in Covid-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in Covid-19 patients with respiratory malfunction and at risk for lung edema. We note that among the currently pursued therapeutic strategies against Covid-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce Covid-19 lethality but will pre-empt a catastrophic scenario in healthcare with insufficient capacity to provide ventilator-assisted respiration.

16.
Neurosci Bull ; 34(1): 120-142, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29282613

RESUMO

Nociception is an important physiological process that detects harmful signals and results in pain perception. In this review, we discuss important experimental evidence involving some TRP ion channels as molecular sensors of chemical, thermal, and mechanical noxious stimuli to evoke the pain and itch sensations. Among them are the TRPA1 channel, members of the vanilloid subfamily (TRPV1, TRPV3, and TRPV4), and finally members of the melastatin group (TRPM2, TRPM3, and TRPM8). Given that pain and itch are pro-survival, evolutionarily-honed protective mechanisms, care has to be exercised when developing inhibitory/modulatory compounds targeting specific pain/itch-TRPs so that physiological protective mechanisms are not disabled to a degree that stimulus-mediated injury can occur. Such events have impeded the development of safe and effective TRPV1-modulating compounds and have diverted substantial resources. A beneficial outcome can be readily accomplished via simple dosing strategies, and also by incorporating medicinal chemistry design features during compound design and synthesis. Beyond clinical use, where compounds that target more than one channel might have a place and possibly have advantageous features, highly specific and high-potency compounds will be helpful in mechanistic discovery at the structure-function level.


Assuntos
Dor/metabolismo , Prurido/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos
18.
Sci Rep ; 8(1): 4878, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29559678

RESUMO

Transient receptor potential vanilloid 4 (TRPV4) cation channels are functional in all renal vascular segments and mediate endothelium-dependent vasorelaxation. Moreover, they are expressed in distinct parts of the tubular system and activated by cell swelling. Ischaemia/reperfusion injury (IRI) is characterized by tubular injury and endothelial dysfunction. Therefore, we hypothesised a putative organ protective role of TRPV4 in acute renal IRI. IRI was induced in TRPV4 deficient (Trpv4 KO) and wild-type (WT) control mice by clipping the left renal pedicle after right-sided nephrectomy. Serum creatinine level was higher in Trpv4 KO mice 6 and 24 hours after ischaemia compared to WT mice. Detailed histological analysis revealed that IRI caused aggravated renal tubular damage in Trpv4 KO mice, especially in the renal cortex. Immunohistological and functional assessment confirmed TRPV4 expression in proximal tubular cells. Furthermore, the tubular damage could be attributed to enhanced necrosis rather than apoptosis. Surprisingly, the percentage of infiltrating granulocytes and macrophages were comparable in IRI-damaged kidneys of Trpv4 KO and WT mice. The present results suggest a renoprotective role of TRPV4 during acute renal IRI. Further studies using cell-specific TRPV4 deficient mice are needed to clarify cellular mechanisms of TRPV4 in IRI.


Assuntos
Túbulos Renais/metabolismo , Traumatismo por Reperfusão/metabolismo , Canais de Cátion TRPV/deficiência , Injúria Renal Aguda/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Isquemia/patologia , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Reperfusão/métodos , Traumatismo por Reperfusão/genética , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
19.
Sci Rep ; 6: 26894, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27247148

RESUMO

TRPV4 ion channels represent osmo-mechano-TRP channels with pleiotropic function and wide-spread expression. One of the critical functions of TRPV4 in this spectrum is its involvement in pain and inflammation. However, few small-molecule inhibitors of TRPV4 are available. Here we developed TRPV4-inhibitory molecules based on modifications of a known TRPV4-selective tool-compound, GSK205. We not only increased TRPV4-inhibitory potency, but surprisingly also generated two compounds that potently co-inhibit TRPA1, known to function as chemical sensor of noxious and irritant signaling. We demonstrate TRPV4 inhibition by these compounds in primary cells with known TRPV4 expression - articular chondrocytes and astrocytes. Importantly, our novel compounds attenuate pain behavior in a trigeminal irritant pain model that is known to rely on TRPV4 and TRPA1. Furthermore, our novel dual-channel blocker inhibited inflammation and pain-associated behavior in a model of acute pancreatitis - known to also rely on TRPV4 and TRPA1. Our results illustrate proof of a novel concept inherent in our prototype compounds of a drug that targets two functionally-related TRP channels, and thus can be used to combat isoforms of pain and inflammation in-vivo that involve more than one TRP channel. This approach could provide a novel paradigm for treating other relevant health conditions.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Dor/tratamento farmacológico , Pancreatite Necrosante Aguda/tratamento farmacológico , Canal de Cátion TRPA1/antagonistas & inibidores , Canais de Cátion TRPV/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Anti-Inflamatórios não Esteroides/síntese química , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Ceruletídeo , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Dor/metabolismo , Dor/fisiopatologia , Pancreatite Necrosante Aguda/induzido quimicamente , Pancreatite Necrosante Aguda/metabolismo , Pancreatite Necrosante Aguda/fisiopatologia , Cultura Primária de Células , Ratos , Suínos , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/metabolismo , Tiazóis/síntese química , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/fisiopatologia
20.
Nat Commun ; 5: 4734, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25178952

RESUMO

Animals need to sense and react to potentially dangerous environments. TRP ion channels participate in nociception, presumably via Ca(2+) influx, in most animal species. However, the relationship between ion permeation and animals' nocifensive behaviour is unknown. Here we use an invertebrate animal model with relevance for mammalian pain. We analyse the putative selectivity filter of OSM-9, a TRPV channel, in osmotic avoidance behaviour of Caenorhabditis elegans. Using mutagenized OSM-9 expressed in the head nociceptor neuron, ASH, we study nocifensive behaviour and Ca(2+) influx. Within the selectivity filter, M(601)-F(609), Y604G strongly reduces avoidance behaviour and eliminates Ca(2+) transients. Y604F also abolishes Ca(2+) transients in ASH, while sustaining avoidance behaviour, yet it disrupts behavioral plasticity. Homology modelling of the OSM-9 pore suggests that Y(604) may assume a scaffolding role. Thus, aromatic residues in the OSM-9 selectivity filter are critical for pain behaviour and ion permeation. These findings have relevance for understanding evolutionary roots of mammalian nociception.


Assuntos
Aprendizagem da Esquiva/fisiologia , Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/fisiologia , Cálcio/metabolismo , Proteínas do Tecido Nervoso/química , Nociceptividade/fisiologia , Nociceptores/metabolismo , Canais de Cátion TRPV/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sinalização do Cálcio , Expressão Gênica , Transporte de Íons , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nociceptores/citologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa