Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ArXiv ; 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38827454

RESUMO

Biological systems, particularly the brain, are frequently analyzed as networks, conveying mechanistic insights into their function and pathophysiology. This is the first study of a functional network of cardiac tissue. We use calcium imaging to obtain two functional networks in a subsidiary but essential pacemaker of the heart, the atrioventricular node (AVN). The AVN is a small cellular structure with dual functions: a) to delay the pacemaker signal passing from the sinoatrial node (SAN) to the ventricles, and b) to serve as a back-up pacemaker should the primary SAN pacemaker fail. Failure of the AVN can lead to syncope and death. We found that the shortest path lengths and clustering coefficients of the AVN are remarkably similar to those of the brain. The network is ``small-world," thus optimized for energy use vs transmission efficiency. We further study the network properties of AVN tissue with knock-out of the sodium-calcium exchange transporter. In this case, the average shortest path-lengths remained nearly unchanged showing network resilience, while the clustering coefficient was somewhat reduced, similar to schizophrenia in brain networks. When we removed the global action potential using principal component analysis (PCA) in wild-type model, the network lost its ``small-world" characteristics with less information-passing efficiency due to longer shortest path lengths but more robust signal propagation resulting from higher clustering. These two wild-type networks (with and without global action potential) may correspond to fast and slow conduction pathways. Laslty, a one-parameter non-linear preferential attachment model is a good fit to all three AVN networks.

2.
J Drugs Dermatol ; 23(3): 160-167, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38443131

RESUMO

Acne vulgaris (AV) is one of the top concerns dermatologists encounter from women. Until now, therapies addressing AV have largely centered around, and have been successful at, targeting the pathophysiological mechanisms that occur at the pilosebaceous unit: sebum hypersecretion, follicular keratinization, over-proliferation of Cutibacterium acnes, and a localized immune response. In addition to these, there is good evidence to suggest that other systemic drivers of a generalized inflammatory response may contribute to the development or exacerbation of acne and that addressing these underlying factors may open more opportunities for developing effective treatments. These include psycho-emotional stress, diet and metabolism, hormonal fluctuations, skin and gut microbiome, oxidative stress, and immune response. While there is accumulating evidence that vitamins, minerals, and botanicals may mitigate some of the pro-inflammatory effects from the activation of these underlying systems, their use and recommendations are limited by a lack of quality efficacy and safety evidence. Here, we present the current evidence for the use of individual supplements in addressing the 6 systemic underlying drivers of AV. We also present a clinical study on the safety and efficacy of a nutraceutical combining many of these ingredients in the management of AV in men and women.  J Drugs Dermatol. 2024;23(3):160-167     doi:10.36849/JDD.8138.


Assuntos
Acne Vulgar , Microbioma Gastrointestinal , Masculino , Feminino , Humanos , Acne Vulgar/tratamento farmacológico , Suplementos Nutricionais , Pele , Vitaminas/uso terapêutico
3.
J Drugs Dermatol ; 23(2): 90-96, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38306144

RESUMO

Acne Vulgaris (AV) is a prominent skin disease commonly affecting teenagers. It often persists into adulthood and is associated with adverse physical and psychosocial impacts. The pathophysiology of AV is conventionally correlated with 4 factors within and around the pilosebaceous unit: increased sebum production, follicular hyperkeratinization, Cutibacterium acnes proliferation, and localized immune responses. As such, conventional therapeutic approaches for AV have primarily focused on these factors. In addition to this primarily localized pathophysiology, there is a progressively emerging body of evidence indicating that underlying systemic factors contributing to a generalized immuno-inflammatory response can contribute to or exacerbate AV. In this article, we introduce and provide the supporting data, for 6 patient-centric systems that may be implicated in the development of AV: psycho-emotional stress, diet and metabolism, dysbiosis of the gut and skin microbiome, hormonal fluctuations, oxidative stress, and immune response. Identifying these pathways and their contributions in a patient-centric approach may provide expanded therapeutic opportunities for treating patients with AV. J Drugs Dermatol. 2024;23(2):90-96.   doi:10.36849/JDD.8137.


Assuntos
Acne Vulgar , Microbiota , Adolescente , Humanos , Acne Vulgar/tratamento farmacológico , Pele/microbiologia , Sebo/metabolismo , Inflamação
5.
Compr Physiol ; 12(1): 2681-2717, 2021 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-34964124

RESUMO

Ca2+ homeostasis is essential for cell function and survival. As such, the cytosolic Ca2+ concentration is tightly controlled by a wide number of specialized Ca2+ handling proteins. One among them is the Na+ -Ca2+ exchanger (NCX), a ubiquitous plasma membrane transporter that exploits the electrochemical gradient of Na+ to drive Ca2+ out of the cell, against its concentration gradient. In this critical role, this secondary transporter guides vital physiological processes such as Ca2+ homeostasis, muscle contraction, bone formation, and memory to name a few. Herein, we review the progress made in recent years about the structure of the mammalian NCX and how it relates to function. Particular emphasis will be given to the mammalian cardiac isoform, NCX1.1, due to the extensive studies conducted on this protein. Given the degree of conservation among the eukaryotic exchangers, the information highlighted herein will provide a foundation for our understanding of this transporter family. We will discuss gene structure, alternative splicing, topology, regulatory mechanisms, and NCX's functional role on cardiac physiology. Throughout this article, we will attempt to highlight important milestones in the field and controversial topics where future studies are required. © 2021 American Physiological Society. Compr Physiol 12:1-37, 2021.


Assuntos
Cálcio , Trocador de Sódio e Cálcio , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Coração , Homeostase , Humanos , Mamíferos/metabolismo , Proteínas de Membrana Transportadoras , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo
6.
J Am Heart Assoc ; 10(17): e019273, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34472363

RESUMO

Background Sodium-calcium (Ca2+) exchanger isoform 1 (NCX1) is the dominant Ca2+ efflux mechanism in cardiomyocytes and is critical to maintaining Ca2+ homeostasis during excitation-contraction coupling. NCX1 activity has been implicated in the pathogenesis of cardiovascular diseases, but a lack of specific NCX1 blockers complicates experimental interpretation. Our aim was to develop a tamoxifen-inducible NCX1 knockout (KO) mouse to investigate compensatory adaptations of acute ablation of NCX1 on excitation-contraction coupling and intracellular Ca2+ regulation, and to examine whether acute KO of NCX1 confers resistance to triggered arrhythmia and ischemia/reperfusion injury. Methods and Results We used the α-myosin heavy chain promoter (Myh6)-MerCreMer promoter to create a tamoxifen-inducible cardiac-specific NCX1 KO mouse. Within 1 week of tamoxifen injection, NCX1 protein expression and current were dramatically reduced. Diastolic Ca2+ increased despite adaptive reductions in Ca2+ current and action potential duration and compensatory increases in excitation-contraction coupling gain, sarcoplasmic reticulum Ca2+ ATPase 2 and plasma membrane Ca2+ ATPase. As these adaptations progressed over 4 weeks, diastolic Ca2+ normalized and SR Ca2+ load increased. Left ventricular function remained normal, but mild fibrosis and hypertrophy developed. Transcriptomics revealed modification of cardiovascular-related gene networks including cell growth and fibrosis. NCX1 KO reduced spontaneous action potentials triggered by delayed afterdepolarizations and reduced scar size in response to ischemia/reperfusion. Conclusions Tamoxifen-inducible NCX1 KO mice adapt to acute genetic ablation of NCX1 by reducing Ca2+ influx, increasing alternative Ca2+ efflux pathways, and increasing excitation-contraction coupling gain to maintain contractility at the cost of mild Ca2+-activated hypertrophy and fibrosis and decreased survival. Nevertheless, KO myocytes are protected against spontaneous action potentials and ischemia/reperfusion injury.


Assuntos
Arritmias Cardíacas , Cálcio , Miócitos Cardíacos , Traumatismo por Reperfusão , Trocador de Sódio e Cálcio , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/prevenção & controle , Cálcio/metabolismo , Fibrose , Hipertrofia , Camundongos , Camundongos Knockout , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Trocador de Sódio e Cálcio/genética , Tamoxifeno/farmacologia
7.
Front Pharmacol ; 11: 903, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595512

RESUMO

Neuronal signals are processed along the nociceptive pathway to convey discriminative information, which would manifest in the produced pain sensation. The transient receptor potential vanilloid 1 (TRPV1), an important signaling complex in nociceptors termini, is activated by different noxious stimuli that underlie distinct pain sensations. For example, while endovanilloids are associated with inflammatory pain and hypersensitivity through TRPV1 activation, the exovanilloid toxin, capsaicin, evokes an acute pain by activating this channel. Differences in the TRPV1 activation profile evoked by exogenous and endogenous vanilloids were suggested to underlie this disparity in pain sensations. However, the cellular processes that lead to these differences in pain sensation mediated by the same channel are not fully understood. Here, we sought to describe the neuronal response of TRPV1-expressing nociceptors to exo-and endovanilloids. To this end, we performed current-clamp recordings in rat trigeminal neurons exposed to either capsaicin or intracellular endovanilloids produced downstream of the bradykinin receptor BK2. Our results show that lipoxygenase metabolites generate persistent TRPV1-dependent action potential firing while capsaicin evokes robust depolarization and high-frequency firing that is quickly terminated by depolarization block. Additionally, we found that a weak TRPV1 activation prolongs action potential firing. Overall, our results indicate different firing patterns evoked by inflammatory mediators and capsaicin via TRPV1 that correlate with the respective subsequent pain sensation. These findings also suggest that differences in neuronal activation stem from the variable degree of TRPV1 activation they produce.

8.
Cell Calcium ; 87: 102167, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32028091

RESUMO

Na/Ca exchange is the dominant calcium (Ca) efflux mechanism in cardiac myocytes. Although our knowledge of exchanger function (NCX1 in the heart) was originally established using biochemical and electrophysiological tools such as cardiac sarcolemmal vesicles and the giant patch technique [1-4], many advances in our understanding of the physiological/pathophysiological roles of NCX1 in the heart have been obtained using a suite of genetically modified mice. Early mouse studies focused on modification of expression levels of NCX1 in the ventricles, with transgenic overexpressors, global NCX1 knockout (KO) mice (which were embryonic lethal if homozygous), and finally ventricular-specific NCX1 KO [5-12]. We found, to our surprise, that ventricular cardiomyocytes lacking NCX1 can survive and function by engaging a clever set of adaptations to minimize Ca entry, while maintaining contractile function through an increase in excitation-contraction (EC) coupling gain [5,6,13]. Having studied ventricular NCX1 ablation in detail, we more recently focused on elucidating the role of NCX1 in the atria through altering NCX1 expression. Using a novel atrial-specific NCX1 KO mouse, we found unexpected changes in atrial cell morphology and calcium handling, together with dramatic alterations in the function of sinoatrial node (SAN) pacemaker activity. In this review, we will discuss these findings and their implications for cardiac disease.


Assuntos
Relógios Biológicos , Cálcio/metabolismo , Acoplamento Excitação-Contração , Átrios do Coração/metabolismo , Nó Sinoatrial/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Sódio/metabolismo , Animais , Humanos
9.
Diabetes ; 67(3): 437-447, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29246974

RESUMO

Polymorphism in TCF7L2, a component of the canonical Wnt signaling pathway, has a strong association with ß-cell dysfunction and type 2 diabetes through a mechanism that has yet to be defined. ß-Cells rely on cells in their microenvironment, including pericytes, for their proper function. Here, we show that Tcf7l2 activity in pancreatic pericytes is required for ß-cell function. Transgenic mice in which Tcf7l2 was selectively inactivated in their pancreatic pericytes exhibited impaired glucose tolerance due to compromised ß-cell function and glucose-stimulated insulin secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of genes required for ß-cell function and maturity in isolated islets. In addition, we identified Tcf7l2-dependent pericytic expression of secreted factors shown to promote ß-cell function, including bone morphogenetic protein 4 (BMP4). Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucose-stimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity affects ß-cells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2-dependent manner to support ß-cell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes and implicates abnormalities in the islet microenvironment in this disease.


Assuntos
Comunicação Celular , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Pericitos/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/uso terapêutico , Diferenciação Celular , Microambiente Celular , Glucose/metabolismo , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Intolerância à Glucose/fisiopatologia , Secreção de Insulina , Células Secretoras de Insulina/patologia , Ligantes , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos Transgênicos , Mutação , Pericitos/patologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Técnicas de Cultura de Tecidos , Proteína 2 Semelhante ao Fator 7 de Transcrição/química , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética
10.
Toxins (Basel) ; 9(10)2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29035314

RESUMO

Beyond providing evolutionary advantages, venoms offer unique research tools, as they were developed to target functionally important proteins and pathways. As a key pain receptor in the nociceptive pathway, transient receptor potential vanilloid 1 (TRPV1) of the TRP superfamily has been shown to be a target for several toxins, as a way of producing pain to deter predators. Importantly, TRPV1 is involved in thermoregulation, inflammation, and acute nociception. As such, toxins provide tools to understand TRPV1 activation and modulation, a critical step in advancing pain research and the development of novel analgesics. Indeed, the phytotoxin capsaicin, which is the spicy chemical in chili peppers, was invaluable in the original cloning and characterization of TRPV1. The unique properties of each subsequently characterized toxin have continued to advance our understanding of functional, structural, and biophysical characteristics of TRPV1. By building on previous reviews, this work aims to provide a comprehensive summary of the advancements made in TRPV1 research in recent years by employing animal toxins, in particular DkTx, RhTx, BmP01, Echis coloratus toxins, APHCs and HCRG21. We examine each toxin's functional aspects, behavioral effects, and structural features, all of which have contributed to our current knowledge of TRPV1. We additionally discuss the key features of TRPV1's outer pore domain, which proves to be the target of the currently discussed toxins.


Assuntos
Canais de Cátion TRPV/efeitos dos fármacos , Toxinas Biológicas/toxicidade , Animais , Venenos de Escorpião/toxicidade , Anêmonas-do-Mar/patogenicidade , Venenos de Serpentes/toxicidade , Venenos de Aranha/toxicidade , Canais de Cátion TRPV/fisiologia
11.
J Vis Exp ; (120)2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28287552

RESUMO

Transfection, the delivery of foreign nucleic acids into a cell, is a powerful tool in protein research. Through this method, ion channels can be investigated through electrophysiological analysis, biochemical characterization, mutational studies, and their effects on cellular processes. Transient transfections offer a simple protocol in which the protein becomes available for analysis within a few hours to days. Although this method presents a relatively straightforward and time efficient protocol, one of the critical components is calibrating the expression of the gene of interest to physiological relevant levels or levels that are suitable for analysis. To this end, many different approaches that offer the ability to control the expression of the gene of interest have emerged. Several stable cell transfection protocols provide a way to permanently introduce a gene of interest into the cellular genome under the regulation of a tetracycline-controlled transcriptional activation. While this technique produces reliable expression levels, each gene of interest requires a few weeks of skilled work including calibration of a killing curve, selection of cell colonies, and overall more resources. Here we present a protocol that uses transient transfection of the Transient Receptor Potential cation channel subfamily V member 1 (TRPV1) gene in an inducible system as an efficient way to express a protein in a controlled manner which is essential in ion channel analysis. We demonstrate that using this technique, we are able to perform calcium imaging, whole cell, and single channel analysis with controlled channel levels required for each type of data collection with a single transfection. Overall, this provides a replicable technique that can be used to study ion channels structure and function.


Assuntos
Regulação da Expressão Gênica , Canais de Cátion TRPV/biossíntese , Transfecção/métodos , Cálcio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Humanos , Plasmídeos/genética , Canais de Cátion TRPV/fisiologia , Tetraciclina/farmacologia
12.
FASEB J ; 31(3): 1238-1247, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27986808

RESUMO

Peripheral neuronal activation by inflammatory mediators is a multifaceted physiological response that involves a multitude of regulated cellular functions. One key pathway that has been shown to be involved in inflammatory pain is Gq/GPCR, whose activation by inflammatory mediators is followed by the regulated response of the cation channel transient receptor potential vanilloid 1 (TRPV1). However, the mechanism that underlies TRPV1 activation downstream of the Gq/GPCR pathway has yet to be fully defined. In this study, we employ pharmacological and molecular biology tools to dissect this activation mechanism via perforated-patch recordings and calcium imaging of both neurons and a heterologous system. We showed that TRPV1 activity downstream of Gq/GPCR activation only produced a subdued current, which was noticeably different from the robust current that is typical of TRPV1 activation by exogenous stimuli. Moreover, we specifically demonstrated that 2 pathways downstream of Gq/GPCR signaling, namely endovanilloid production by lipoxygenases and channel phosphorylation by PKC, converge on TRPV1 to evoke a tightly regulated response. Of importance, we show that only when both pathways are acting on TRPV1 is the inflammatory-mediated response achieved. We propose that the requirement of multiple signaling events allows subdued TRPV1 activation to evoke regulated neuronal response during inflammation.-Kumar R., Hazan, A., Geron, M., Steinberg, R., Livni, L., Matzner, H., Priel, A. Activation of transient receptor potential vanilloid 1 by lipoxygenase metabolites depends on PKC phosphorylation.


Assuntos
Lipoxigenase/metabolismo , Proteína Quinase C/metabolismo , Canais de Cátion TRPV/metabolismo , Potenciais de Ação , Animais , Células Cultivadas , Eicosanoides/farmacologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos
13.
J Biol Chem ; 291(26): 13855-63, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27143360

RESUMO

Vanilloids are pain evoking molecules that serve as ligands of the "heat and capsaicin receptor" TRPV1. Binding of either endogenous or exogenous vanilloids evokes channel and subsequent neuronal activation, leading to pain sensation. Despite its pivotal physiological role, the molecular basis of TRPV1 activation and deactivation is not fully understood. The highly conserved tyrosine in position 511 (Tyr(511)) of the rat TRPV1 (rTRPV1) was the first residue to be identified as a necessary participant in the vanilloid-mediated response. rTRPV1 cryo-EM structures implicated rotation of this residue in the vanilloids bound state. Therefore, we hypothesize that the rTRPV1 Tyr(511) residue entraps vanilloids in their binding site, prolonging channel activity. To test our hypothesis, we generated an array of rTRPV1 mutants, containing the whole spectrum of Tyr(511) substitutions, and tested their response to both exo- and endovanilloids. Our data show that only substitutions of Tyr(511) to aromatic amino acids were able to mimic, albeit partially, the vanilloid-evoked activation pattern of the wt receptor. Although these substitutions reduced the channel sensitivity to vanilloids, a maximal open-channel lifetime could be achieved. Moreover, whereas their current activation rate remains intact, receptors with Tyr(511) substitutions exhibited a faster current deactivation. Our findings therefore suggest that the duration of channel activity evoked by vanilloids is regulated by the interaction between Tyr(511) and the agonist. To conclude, we suggest that Tyr(511)-mediated anchoring of vanilloids in their binding pocket is pivotal for TRPV1 activation and subsequent pain sensation.


Assuntos
Canais de Cátion TRPV/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Cinética , Mutação de Sentido Incorreto , Dor/genética , Dor/metabolismo , Ratos , Canais de Cátion TRPV/genética , Tirosina
14.
Sci Rep ; 5: 12278, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26194846

RESUMO

The receptor channel TRPV1 (Transient Receptor Potential Vanilloid 1) is expressed by primary afferent sensory neurons of the pain pathway, where it functions as a sensor of noxious heat and various chemicals, including eicosanoids, capsaicin, protons and peptide toxins. Comprised of four identical subunits that organize into a non-selective cationic permeable channel, this receptor has a variety of binding sites responsible for detecting their respective agonists. Although its physiological role as a chemosensor has been described in detail, the stoichiometry of TRPV1 activation by its different ligands remains unknown. Here, we combined the use of concatemeric constructs harboring mutated binding sites with patch-clamp recordings in order to determine the stoichiometry for TRPV1 activation through the vanilloid binding site and the outer-pore domain by capsaicin and protons, respectively. We show that, while a single capsaicin-bound subunit was sufficient to achieve a maximal open-channel lifetime, all four proton-binding sites were required. Thus, our results demonstrate a distinct stoichiometry of TRPV1 activation through two of its different agonist-binding domains.


Assuntos
Canais de Cátion TRPV/agonistas , Animais , Sítios de Ligação , Capsaicina/farmacologia , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Prótons , Ratos , Canais de Cátion TRPV/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...