Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Biosens Bioelectron ; 241: 115668, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37774465

RESUMO

Continuous glucose monitoring schemes that avoid finger pricking are of utmost importance to enhance the comfort and lifestyle of diabetic patients. To this aim, we propose a microwave planar sensing platform as a potent sensing technology that extends its applications to biomedical analytes. In this paper, a compact planar resonator-based sensor is introduced for noncontact sensing of glucose. Furthermore, in vivo and in-vitro tests using a microfluidic channel system and in clinical trial settings demonstrate its reliable operation. The proposed sensor offers real-time response and a high linear correlation (R2 ∼ 0.913) between the measured sensor response and the blood glucose level (GL). The sensor is also enhanced with machine learning to predict the variation of body glucose levels for non-diabetic and diabetic patients. This addition is instrumental in triggering preemptive measures in cases of unusual glucose level trends. In addition, it allows for the detection of common artifacts of the sensor as anomalies so that they can be removed from the measured data. The proposed system is designed to noninvasively monitor interstitial glucose levels in humans, introducing the opportunity to create a customized wearable apparatus with the ability to learn.


Assuntos
Técnicas Biossensoriais , Diabetes Mellitus , Humanos , Glicemia , Automonitorização da Glicemia , Micro-Ondas , Glucose , Diabetes Mellitus/diagnóstico , Aprendizado de Máquina
2.
J Physiol ; 601(1): 83-98, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36420836

RESUMO

Autosomal dominant polycystic kidney disease is caused by mutations in the membrane receptor PKD1 or the cation channel PKD2. TACAN (also termed TMEM120A), recently reported as an ion channel in neurons for mechanosensing and pain sensing, is also distributed in diverse non-neuronal tissues, such as kidney, heart and intestine, suggesting its involvement in other functions. In this study, we found that TACAN is in a complex with PKD2 in native renal cell lines. Using the two-electrode voltage clamp in Xenopus oocytes, we found that TACAN inhibits the channel activity of PKD2 gain-of-function mutant F604P. TACAN fragments containing the first and last transmembrane domains interacted with the PKD2 C- and N-terminal fragments, respectively. The TACAN N-terminus acted as a blocking peptide, and TACAN inhibited the function of PKD2 by the binding of PKD2 with TACAN. By patch clamping in mammalian cells, we found that TACAN inhibits both the single-channel conductance and the open probability of PKD2 and mutant F604P. PKD2 co-expressed with TACAN, but not PKD2 alone, exhibited pressure sensitivity. Furthermore, we found that TACAN aggravates PKD2-dependent tail curvature and pronephric cysts in larval zebrafish. In summary, this study revealed that TACAN acts as a PKD2 inhibitor and mediates mechanosensitivity of the PKD2-TACAN channel complex. KEY POINTS: TACAN inhibits the function of PKD2 in vitro and in vivo. TACAN N-terminal S1-containing fragment T160X interacts with the PKD2 C-terminal fragment N580-L700, and its C-terminal S6-containing fragment L296-D343 interacts with the PKD2 N-terminal A594X. TACAN inhibits the function of the PKD2 channel by physical interaction. The complex of PKD2 with TACAN, but not PKD2 alone, confers mechanosensitivity.


Assuntos
Rim Policístico Autossômico Dominante , Peixe-Zebra , Animais , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Canais Iônicos/genética , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Rim/metabolismo , Mamíferos/metabolismo
3.
IEEE Trans Biomed Eng ; 69(10): 3175-3182, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35333709

RESUMO

Monitoring lactate levels is an established method for determining hyperlactatemia in critically ill patients and assessing aerobic fitness. It is a widely used gold-standard technique in both professional and serious amateur sports. Non-invasive real-time lactate monitoring offers significant advantages over the current technology of finger-prick blood sampling. Possible candidate technology for developing non-invasive real-time lactate monitoring should be highly sensitive, flexible, and capable of real-time monitoring of lactate levels in interstitial fluid or within specific working muscle groups depending on the type of sport. Herein we describe a planar, flexible, passive, chipless tag resonator that is electromagnetically coupled to a reader placed in proximity to the lactate sensor tag. The tag resonator is a thin metallic tracing that can be taped on the skin. The resonance frequency of the tag fluctuates proportionately with changing lactate concentrations in a solution mimicking human interstitial fluid with very high sensitivity. The spectrum of the tag is reflected in the spectrum of the reader, which is a planar microwave resonator designed at a different frequency. The reader could be embedded in a cellphone or an application-specific wearable device for data communication and processing. The tag can accurately and reproducibly measure lactate concentrations in the range of 1 to 10 mM, which is in the physiological range of lactate observed at rest and during intense physical activity. Furthermore, the chrematistics of this technology will allow monitoring of lactate in specific working muscle groups.


Assuntos
Ácido Láctico , Dispositivos Eletrônicos Vestíveis , Líquido Extracelular , Humanos , Micro-Ondas , Monitorização Fisiológica
4.
J Mol Cell Cardiol ; 167: 17-31, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35331696

RESUMO

Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute a promising drug treatment for heart failure patients with either preserved or reduced ejection fraction. Whereas SGLT2i were originally developed to target SGLT2 in the kidney to facilitate glucosuria in diabetic patients, it is becoming increasingly clear that these drugs also have important effects outside of the kidney. In this review we summarize the literature on cardiac effects of SGLT2i, focussing on pro-inflammatory and oxidative stress processes, ion transport mechanisms controlling sodium and calcium homeostasis and metabolic/mitochondrial pathways. These mechanisms are particularly important as disturbances in these pathways result in endothelial dysfunction, diastolic dysfunction, cardiac stiffness, and cardiac arrhythmias that together contribute to heart failure. We review the findings that support the concept that SGLT2i directly and beneficially interfere with inflammation, oxidative stress, ionic homeostasis, and metabolism within the cardiac cell. However, given the very low levels of SGLT2 in cardiac cells, the evidence suggests that SGLT2-independent effects of this class of drugs likely occurs via off-target effects in the myocardium. Thus, while there is still much to be understood about the various factors which determine how SGLT2i affect cardiac cells, much of the research clearly demonstrates that direct cardiac effects of these SGLT2i exist, albeit mediated via SGLT2-independent pathways, and these pathways may play a role in explaining the beneficial effects of SGLT2 inhibitors in heart failure.


Assuntos
Diabetes Mellitus Tipo 2 , Insuficiência Cardíaca , Inibidores do Transportador 2 de Sódio-Glicose , Humanos , Miocárdio/metabolismo , Transportador 2 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/uso terapêutico , Inibidores do Transportador 2 de Sódio-Glicose/efeitos adversos
5.
Cell Metab ; 34(2): 256-268.e5, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108513

RESUMO

In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "ß cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagon , Ilhotas Pancreáticas , Animais , Diabetes Mellitus Tipo 2/metabolismo , Exocitose/fisiologia , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos
8.
Circulation ; 143(22): 2188-2204, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33832341

RESUMO

BACKGROUND: SGLT2 (sodium/glucose cotransporter 2) inhibitors exert robust cardioprotective effects against heart failure in patients with diabetes, and there is intense interest to identify the underlying molecular mechanisms that afford this protection. Because the induction of the late component of the cardiac sodium channel current (late-INa) is involved in the etiology of heart failure, we investigated whether these drugs inhibit late-INa. METHODS: Electrophysiological, in silico molecular docking, molecular, calcium imaging, and whole heart perfusion techniques were used to address this question. RESULTS: The SGLT2 inhibitor empagliflozin reduced late-INa in cardiomyocytes from mice with heart failure and in cardiac Nav1.5 sodium channels containing the long QT syndrome 3 mutations R1623Q or ΔKPQ. Empagliflozin, dapagliflozin, and canagliflozin are all potent and selective inhibitors of H2O2-induced late-INa (half maximal inhibitory concentration = 0.79, 0.58, and 1.26 µM, respectively) with little effect on peak sodium current. In mouse cardiomyocytes, empagliflozin reduced the incidence of spontaneous calcium transients induced by the late-INa activator veratridine in a similar manner to tetrodotoxin, ranolazine, and lidocaine. The putative binding sites for empagliflozin within Nav1.5 were investigated by simulations of empagliflozin docking to a three-dimensional homology model of human Nav1.5 and point mutagenic approaches. Our results indicate that empagliflozin binds to Nav1.5 in the same region as local anesthetics and ranolazine. In an acute model of myocardial injury, perfusion of isolated mouse hearts with empagliflozin or tetrodotoxin prevented activation of the cardiac NLRP3 (nuclear-binding domain-like receptor 3) inflammasome and improved functional recovery after ischemia. CONCLUSIONS: Our results provide evidence that late-INa may be an important molecular target in the heart for the SGLT2 inhibitors, contributing to their unexpected cardioprotective effects.


Assuntos
Compostos Benzidrílicos/farmacologia , Glucosídeos/farmacologia , Canais de Sódio/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Animais , Compostos Benzidrílicos/uso terapêutico , Glucosídeos/uso terapêutico , Humanos , Masculino , Camundongos , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico
9.
Islets ; 13(1-2): 32-50, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33724156

RESUMO

Glucagon-Like Peptide-1 (GLP-1) is an important peptide hormone secreted by L-cells in the gastrointestinal tract in response to nutrients. It is produced by the differential cleavage of the proglucagon peptide. GLP-1 elicits a wide variety of physiological responses in many tissues that contribute to metabolic homeostasis. For these reasons, therapies designed to either increase endogenous GLP-1 levels or introduce exogenous peptide mimetics are now widely used in the management of diabetes. In addition to GLP-1 production from L-cells, recent reports suggest that pancreatic islet alpha cells may also synthesize and secrete GLP-1. Intra-islet GLP-1 may therefore play an unappreciated role in islet health and glucose regulation, suggesting a potential functional paracrine role for islet-derived GLP-1. In this review, we assess the current literature from an islet-centric point-of-view to better understand the production, degradation, and actions of GLP-1 within the endocrine pancreas in rodents and humans. The relevance of intra-islet GLP-1 in human physiology is discussed regarding the potential role of intra-islet GLP-1 in islet health and dysfunction.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/fisiologia , Células Secretoras de Glucagon , Ilhotas Pancreáticas , Glucagon , Glucose , Humanos
10.
PLoS One ; 15(8): e0236457, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32790682

RESUMO

Transgenic mice with selective induction of calreticulin transgene expression in cardiomyocytes (CardiacCRT+) were analyzed. CardiacCRT+ cardiomyocytes showed increased contractility and Ca2+ transients. Yet, in vivo assessment of cardiac performance, and ischemic tolerance of CardiacCRT+ mice demonstrated right ventricle dilation and reduced cardiac output, increased QT interval and decreased P amplitude. Paradoxically, ex vivo working hearts from CardiacCRT+ mice showed enhanced ischemic cardio-protection and cardiac efficiency. Under aerobic conditions, CardiacCRT+ hearts showed less efficient cardiac function than sham control hearts due to an increased ATP production from glycolysis relative to glucose oxidation. During reperfusion, this inefficiency was reversed, with CardiacCRT+ hearts exhibiting better functional recovery and increased cardiac efficiency compared to sham control hearts. On the other hand, mechanical stretching of isolated cardiac fibroblasts activated the IRE1α branch of the unfolded protein response pathway as well as induction of Col1A2 and TGFß gene expression ex vivo, which were all suppressed by tauroursodeoxycholic acid.


Assuntos
Calreticulina/metabolismo , Contração Miocárdica , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Calreticulina/genética , Células Cultivadas , Metabolismo Energético , Frequência Cardíaca , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/patologia , Regulação para Cima
11.
Sci Rep ; 10(1): 12980, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737348

RESUMO

This paper reports a highly sensitive, non-invasive sensor for real-time glucose monitoring from interstitial fluid. The structure is comprised of a chip-less tag sensor which may be taped over the patient's skin and a reader, that can be embedded in a smartwatch. The tag sensor is energized through the established electromagnetic coupling between the tag and the reader and its frequency response is reflected on the spectrum of the reader in the same manner. The tag sensor consumes zero power as there is no requirement for any active readout or communication circuitry on the tag side. When measuring changes in glucose concentrations within saline replicating interstitial fluid, the sensor was able to detect glucose with an accuracy of ~ 1 mM/l over a physiological range of glucose concentrations with 38 kHz of the resonance frequency shift. This high sensitivity is attained as a result of the proposed new design and extended field concentration on the tag. The impact of some of the possible interferences on the response of the sensor's performance was also investigated. Variations in electrolyte concentrations within the test samples have a negligible effect on the response of the sensor unless these variations are supra-physiologically large.

12.
J Physiol ; 598(19): 4321-4338, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32721035

RESUMO

KEY POINTS: 25-Hydroxyvitamin D (25OHD) is a partial agonist of TRPV1 whereby 25OHD can weakly activate TRPV1 yet antagonize the stimulatory effects of the full TRPV1 agonists capsaicin and oleoyl dopamine. 25OHD binds to TRPV1 within the same vanilloid binding pocket as capsaicin. 25OHD inhibits the potentiating effects of PKC-mediated TRPV1 activity. 25OHD reduces T-cell activation and trigeminal neuron calcium signalling mediated by TRPV1 activity. These results provide evidence that TRPV1 is a novel receptor for the biological actions of vitamin D in addition to the well-documented effects of vitamin D upon the nuclear vitamin D receptor. The results may have important implications for our current understanding of certain diseases where TRPV1 and vitamin D deficiency have been implicated, such as chronic pain and autoimmune diseases, such as type 1 diabetes. ABSTRACT: The capsaicin receptor TRPV1 plays an important role in nociception, inflammation and immunity and its activity is regulated by exogenous and endogenous lipophilic ligands. As vitamin D is lipophilic and involved in similar biological processes as TRPV1, we hypothesized that it directly regulates TRPV1 activity and function. Our calcium imaging and electrophysiological data demonstrate that vitamin D (25-hydroxyvitamin D (25OHD) and 1,25-hydroxyvitamin D (1,25OHD)) can weakly activate TRPV1 at physiologically relevant concentrations (100 nM). Furthermore, both 25OHD and 1,25OHD can inhibit capsaicin-induced TRPV1 activity (IC50  = 34.3 ± 0.2 and 11.5 ± 0.9 nM, respectively), but not pH-induced TRPV1 activity, suggesting that vitamin D interacts with TRPV1 in the same region as the TRPV1 agonist capsaicin. This hypothesis is supported by our in silico TRPV1 structural modelling studies, which place 25OHD in the same binding region as capsaicin. 25OHD also attenuates PKC-dependent TRPV1 potentiation via interactions with a known PKC phospho-acceptor residue in TRPV1. To provide evidence for a physiological role for the interaction of vitamin D with TRPV1, we employed two different cellular models known to express TRPV1: mouse CD4+ T-cells and trigeminal neurons. Our results indicate that 25OHD reduces TRPV1-induced cytokine release from T-cells and capsaicin-induced calcium activity in trigeminal neurons. In summary, we provide evidence that vitamin D is a novel endogenous regulator of TRPV1 channel activity that may play an important physiological role in addition to its known effects through the canonical nuclear vitamin D receptor pathway.


Assuntos
Canais de Potencial de Receptor Transitório , Animais , Capsaicina/farmacologia , Camundongos , Neurônios , Ratos Sprague-Dawley , Canais de Cátion TRPV , Vitamina D/farmacologia
13.
Mol Metab ; 39: 101014, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32413586

RESUMO

OBJECTIVES: Our study shows that glucagon-like peptide-1 (GLP-1) is secreted within human islets and may play an unexpectedly important paracrine role in islet physiology and pathophysiology. It is known that α cells within rodent and human pancreatic islets are capable of secreting GLP-1, but little is known about the functional role that islet-derived GLP-1 plays in human islets. METHODS: We used flow cytometry, immunohistochemistry, perifusions, and calcium imaging techniques to analyse GLP-1 expression and function in islets isolated from cadaveric human donors with or without type 2 diabetes. We also used immunohistochemistry to analyse GLP-1 expression within islets from pancreatic biopsies obtained from living donors. RESULTS: We have demonstrated that human islets secrete ∼50-fold more GLP-1 than murine islets and that ∼40% of the total human α cells contain GLP-1. Our results also confirm that dipeptidyl peptidase-4 (DPP4) is expressed in α cells. Sitagliptin increased GLP-1 secretion from cultured human islets but did not enhance glucose-stimulated insulin secretion (GSIS) in islets from non-diabetic (ND) or type 2 diabetic (T2D) donors, suggesting that ß cell GLP-1 receptors (GLP-1R) may already be maximally activated. Therefore, we tested the effects of exendin-9, a GLP-1R antagonist. Exendin-9 was shown to reduce GSIS by 39% and 61% in ND islets and T2D islets, respectively. We also observed significantly more GLP-1+ α cells in T2D islets compared with ND islets obtained from cadaveric donors. Furthermore, GLP-1+ α cells were also identified in pancreatic islet sections obtained from living donors undergoing surgery. CONCLUSIONS: In summary, we demonstrated that human islets secrete robust amounts of GLP-1 from an α cell subpopulation and that GLP-1R signalling may support GSIS to a greater extent in T2D islets.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Células Secretoras de Glucagon/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Biomarcadores , Diabetes Mellitus Tipo 2/etiologia , Expressão Gênica , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/genética , Glucose/metabolismo , Humanos , Imunofenotipagem , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos
14.
Circ Heart Fail ; 13(1): e006277, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31957470

RESUMO

BACKGROUND: Although empagliflozin was shown to profoundly reduce cardiovascular events in diabetic patients and blunt the decline in cardiac function in nondiabetic mice with established heart failure (HF), the mechanism of action remains unknown. METHODS AND RESULTS: We treated 2 rodent models of HF with 10 mg/kg per day empagliflozin and measured activation of the NLRP3 (nucleotide-binding domain-like receptor protein 3) inflammasome in the heart. We show for the first time that beneficial effects of empagliflozin in HF with reduced ejection fraction (HF with reduced ejection fraction [HFrEF]; n=30-34) occur in the absence of changes in circulating ketone bodies, cardiac ketone oxidation, or increased cardiac ATP production. Of note, empagliflozin attenuated activation of the NLRP3 inflammasome and expression of associated markers of sterile inflammation in hearts from mice with HFrEF, implicating reduced cardiac inflammation as a mechanism of empagliflozin that contributes to sustained function in HFrEF in the absence of diabetes mellitus. In addition, we validate that the beneficial cardiac effects of empagliflozin in HF with preserved ejection fraction (HFpEF; n=9-10) are similarly associated with reduced activation of the NLRP3 inflammasome. Lastly, the ability of empagliflozin to reduce inflammation was completely blunted by a calcium (Ca2+) ionophore, suggesting that empagliflozin exerts its benefit upon restoring optimal cytoplasmic Ca2+ levels in the heart. CONCLUSIONS: These data provide evidence that the beneficial cardiac effects of empagliflozin are associated with reduced cardiac inflammation via blunting activation of the NLRP3 inflammasome in a Ca2+-dependent manner and hence may be beneficial in treating HF even in the absence of diabetes mellitus.


Assuntos
Compostos Benzidrílicos/farmacologia , Glucosídeos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Volume Sistólico/efeitos dos fármacos , Animais , Proteínas de Transporte/metabolismo , Cardiopatias/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Nucleotídeos/metabolismo , Volume Sistólico/fisiologia
15.
JCI Insight ; 4(1)2019 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-30626749

RESUMO

Obese individuals are often at risk for nonalcoholic fatty liver disease (NAFLD), insulin resistance, type 2 diabetes (T2D), and cardiovascular diseases such as angina, thereby requiring combination therapies for their comorbidities. Ranolazine is a second-line antianginal agent that also improves glycemia, and our aim was to determine whether ranolazine modifies the progression of obesity-induced NAFLD. Twelve-week-old C57BL/6J male mice were fed a low-fat or high-fat diet for 10 weeks and then treated for 30 days with either vehicle control or ranolazine (50 mg/kg via daily s.c. injection). Glycemia was monitored via glucose/pyruvate/insulin tolerance testing, whereas in vivo metabolism was assessed via indirect calorimetry. Hepatic triacylglycerol content was quantified via the Bligh and Dyer method. Consistent with previous reports, ranolazine treatment reversed obesity-induced glucose intolerance, which was associated with reduced body weight and hepatic steatosis, as well as increased hepatic pyruvate dehydrogenase (PDH) activity. Ranolazine's actions on hepatic PDH activity may be directly mediated, as ranolazine treatment reduced PDH phosphorylation (indicative of increased PDH activity) in HepG2 cells. Therefore, in addition to mitigating angina, ranolazine also reverses NAFLD, which may contribute to its documented glucose-lowering actions, situating ranolazine as an ideal antianginal therapy for obese patients comorbid for NAFLD and T2D.

16.
Cell Rep ; 22(6): 1560-1573, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29425510

RESUMO

Transient receptor potential (TRP) channels are regulated by diverse stimuli comprising thermal, chemical, and mechanical modalities. They are also commonly regulated by phosphatidylinositol-4,5-bisphosphate (PIP2), with underlying mechanisms largely unknown. We here revealed an intramolecular interaction of the TRPP3 N and C termini (N-C) that is functionally essential. The interaction was mediated by aromatic Trp81 in pre-S1 domain and cationic Lys568 in TRP-like domain. Structure-function analyses revealed similar N-C interaction in TRPP2 as well as TRPM8/-V1/-C4 via highly conserved tryptophan and lysine/arginine residues. PIP2 bound to cationic residues in TRPP3, including K568, thereby disrupting the N-C interaction and negatively regulating TRPP3. PIP2 had similar negative effects on TRPP2. Interestingly, we found that PIP2 facilitates the N-C interaction in TRPM8/-V1, resulting in channel potentiation. The intramolecular N-C interaction might represent a shared mechanism underlying the gating and PIP2 regulation of TRP channels.


Assuntos
Ativação do Canal Iônico/fisiologia , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potencial de Receptor Transitório/química , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos , Ligação Proteica , Domínios Proteicos , Relação Estrutura-Atividade , Xenopus laevis
17.
Sci Rep ; 7(1): 16332, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29180820

RESUMO

Subcutaneous white adipose tissue (scWAT) is the major fat depot in humans and is a central player in regulating whole body metabolism. Skin exposure to UV wavelengths from sunlight is required for Vitamin D synthesis and pigmentation, although it is plausible that longer visible wavelengths that penetrate the skin may regulate scWAT function. In this regard, we discovered a novel blue light-sensitive current in human scWAT that is mediated by melanopsin coupled to transient receptor potential canonical cation channels. This pathway is activated at physiological intensities of light that penetrate the skin on a sunny day. Daily exposure of differentiated adipocytes to blue light resulted in decreased lipid droplet size, increased basal lipolytic rate and alterations in adiponectin and leptin secretion. Our results suggest that scWAT function may be directly under the influence of ambient sunlight exposure and may have important implications for our current understanding of adipocyte biology. (150 words).


Assuntos
Adipócitos Brancos/metabolismo , Transdução de Sinal Luminoso , Opsinas de Bastonetes/metabolismo , Canais de Cátion TRPC/metabolismo , Células 3T3-L1 , Adipocinas/biossíntese , Animais , Fenômenos Eletrofisiológicos , Humanos , Luz , Metabolismo dos Lipídeos/efeitos da radiação , Camundongos , Opsinas de Bastonetes/genética , Gordura Subcutânea/citologia , Gordura Subcutânea/metabolismo , Canais de Cátion TRPC/genética
18.
Am J Physiol Heart Circ Physiol ; 313(3): H469-H478, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28667052

RESUMO

Cardiac ATP-sensitive K+ (KATP) channels couple changes in cellular metabolism to membrane excitability and are activated during metabolic stress, although under basal aerobic conditions, KATP channels are thought to be predominately closed. Despite intense research into the roles of KATP channels during metabolic stress, their contribution to aerobic basal cardiac metabolism has not been previously investigated. Hearts from Kir6.2+/+ and Kir6.2-/- mice were perfused in working mode, and rates of glycolysis, fatty acid oxidation, and glucose oxidation were measured. Changes in activation/expression of proteins regulating metabolism were probed by Western blot analysis. Despite cardiac mechanical function and metabolic efficiency being similar in both groups, hearts from Kir6.2-/- mice displayed an approximately twofold increase in fatty acid oxidation and a 0.45-fold reduction in glycolytic rates but similar glucose oxidation rates compared with hearts from Kir6.2+/+ mice. Kir6.2-/- hearts also possessed elevated levels of activated AMP-activated protein kinase (AMPK), higher glycogen content, and reduced mitochondrial density. Moreover, activation of AMPK by isoproterenol or diazoxide was significantly blunted in Kir6.2-/- hearts. These data indicate that KATP channel ablation alters aerobic basal cardiac metabolism. The observed increase in fatty acid oxidation and decreased glycolysis before any metabolic insult may contribute to the poor recovery observed in Kir6.2-/- hearts in response to exercise or ischemia-reperfusion injury. Therefore, KATP channels may play an important role in the regulation of cardiac metabolism through AMPK signaling.NEW & NOTEWORTHY In this study, we show that genetic ablation of plasma membrane ATP-sensitive K+ channels results in pronounced changes in cardiac metabolic substrate preference and AMP-activated protein kinase activity. These results suggest that ATP-sensitive K+ channels may play a novel role in regulating metabolism in addition to their well-documented effects on ionic homeostasis during periods of stress.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Membrana Celular/enzimologia , Metabolismo Energético , Miócitos Cardíacos/enzimologia , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Ácidos Graxos/metabolismo , Genótipo , Glucose/metabolismo , Glicólise , Preparação de Coração Isolado , Cinética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/enzimologia , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/ultraestrutura , Oxirredução , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Fatores de Tempo
19.
Cell Metab ; 25(6): 1334-1347.e4, 2017 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-28591636

RESUMO

The processes contributing to ß cell dysfunction in type 2 diabetes (T2D) are uncertain, largely because it is difficult to access ß cells in their intact immediate environment. We examined the pathophysiology of ß cells under T2D progression directly in pancreatic tissues. We used MALDI imaging of Langerhans islets (LHIs) within mouse tissues or from human tissues to generate in situ-omics data, which we supported with in vitro experiments. Molecular interaction networks provided information on functional pathways and molecules. We found that stearoylcarnitine accumulated in ß cells, leading to arrest of insulin synthesis and energy deficiency via excessive ß-oxidation and depletion of TCA cycle and oxidative phosphorylation metabolites. Acetylcarnitine and an accumulation of N-acyl taurines, a group not previously detected in ß cells, provoked insulin secretion. Thus, ß cell dysfunction results from enhanced insulin secretion combined with an arrest of insulin synthesis.


Assuntos
Carnitina/análogos & derivados , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Taurina/efeitos adversos , Animais , Carnitina/efeitos adversos , Carnitina/farmacologia , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Humanos , Secreção de Insulina , Células Secretoras de Insulina/patologia , Camundongos , Taurina/farmacologia
20.
J Mol Cell Cardiol ; 108: 34-41, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28483598

RESUMO

Cardiac ATP-sensitive K+ (KATP) channel activity plays an important cardio-protective role in regulating excitability in response to metabolic stress. Evidence suggests that these channels are also mechano-sensitive and therefore may couple KATP channel activity to increased cardiac workloads. However, the molecular mechanism that couples membrane stretch to channel activity is not currently known. We hypothesized that membrane stretch may alter the intrinsic MgATPase activity of the cardiac KATP channel resulting in increased channel activation. The inside-out patch-clamp technique was used to record single-channel and macroscopic recombinant KATP channel activity in response to membrane stretch elicited by negative pipette pressure. We found that stretch activation requires the presence of the SUR subunit and that inhibition of MgATPase activity with either the non-hydrolysable ATP analog AMP-PNP or the ATPase inhibitor BeFx significantly reduced the stimulatory effect of stretch. We employed a point mutagenic approach to determine that a single residue (K1337) in the hairpin loop proximal to the major MgATPase catalytic site in the SUR2A subunit is responsible for the difference in mechano-sensitivity between SUR2A and SUR1 containing KATP channels. Moreover, using a double cysteine mutant substitution in the hairpin loop region revealed the importance of a key residue-residue interaction in this region that transduces membrane mechanical forces into KATP channel stimulation via increases in channel MgATPase activity. With respect to KATP channel pharmacology, glibenclamide, but not glicalizide or repaglinide, was able to completely inhibit KATP channel mechano-sensitivity. In summary, our results provide a highly plausible molecular mechanism by which mechanical membrane forces are rapidly converted in changes in KATP channel activity that have implications for our understanding of cardiac KATP channels in physiological or pathophysiological settings that involve increased workload.


Assuntos
Adenosina Trifosfatases/metabolismo , Canais KATP/metabolismo , Mecanotransdução Celular , Contração Miocárdica , Miocárdio/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Sequência de Aminoácidos , Substituição de Aminoácidos , Ativação Enzimática , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico , Canais KATP/química , Canais KATP/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Subunidades Proteicas , Relação Estrutura-Atividade , Receptores de Sulfonilureias/química , Receptores de Sulfonilureias/genética , Receptores de Sulfonilureias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA