Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4915, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38851747

RESUMO

The bioavailability of nicotinamide adenine dinucleotide (NAD) is vital for skeletal muscle health, yet the mechanisms or signals regulating NAD homeostasis remain unclear. Here, we uncover a pathway connecting peripheral glucose sensing to the modulation of muscle NAD through TAS1R2, the sugar-sensing G protein-coupled receptor (GPCR) initially identified in taste perception. Muscle TAS1R2 receptor stimulation by glucose and other agonists induces ERK1/2-dependent phosphorylation and activation of poly(ADP-ribose) polymerase1 (PARP1), a major NAD consumer in skeletal muscle. Consequently, muscle-specific deletion of TAS1R2 (mKO) in male mice suppresses PARP1 activity, elevating NAD levels and enhancing mitochondrial capacity and running endurance. Plasma glucose levels negatively correlate with muscle NAD, and TAS1R2 receptor deficiency enhances NAD responses across the glycemic range, implicating TAS1R2 as a peripheral energy surveyor. These findings underscore the role of GPCR signaling in NAD regulation and propose TAS1R2 as a potential therapeutic target for maintaining muscle health.


Assuntos
Glucose , Homeostase , Músculo Esquelético , NAD , Receptores Acoplados a Proteínas G , Animais , Músculo Esquelético/metabolismo , NAD/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Masculino , Glucose/metabolismo , Camundongos , Camundongos Knockout , Humanos , Mitocôndrias/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fosforilação
2.
Res Sq ; 2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36798161

RESUMO

Muscle fitness and mass deteriorate under the conditions of obesity and aging for reasons yet to be fully elucidated. Herein, we describe a novel pathway linking peripheral nutrient sensing and skeletal muscle function through the sweet taste receptor TAS1R2 and the involvement of ERK2-PARP1-NAD signaling axis. Muscle-specific deletion of TAS1R2 (mKO) in mice produced elevated NAD levels due to suppressed PARP1 activity, improved mitochondrial function, increased muscle mass and strength, and prolonged running endurance. Deletion of TAS1R2 in obese or aged mice also ameliorated the decline in muscle mass and fitness arising from these conditions. Remarkably, partial loss-of-function of TAS1R2 (rs35874116) in older, obese humans recapitulated the healthier muscle phenotype displayed by mKO mice in response to exercise training. Our findings show that inhibition of the TAS1R2 signaling in skeletal muscle is a promising therapeutic approach to preserve muscle mass and function.

3.
Front Nutr ; 9: 896205, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35662939

RESUMO

The Ile191Val variant of the TAS1R2 gene of sweet taste receptors causes a partial loss-of-function and is associated with reduced glucose excursions in a healthy lean cohort. However, it is unclear whether this polymorphism contributes to the regulation of glucose homeostasis in metabolically unhealthy individuals. Thus, we used participants with variable glycemic profiles and obesity to assess the effects of the TAS1R2-Ile191Val variant. We found that the Val minor allele carriers had lower HbA1c at all levels of fasting glucose and glucose tolerance. These effects were not due to differences in beta-cell function or insulin sensitivity assessed with a frequently sampled intravenous glucose tolerance test. This study extends our previous findings and provides further evidence that sweet taste receptor function may contribute to glucose regulation in humans.

4.
Biomedicines ; 10(1)2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35052799

RESUMO

BACKGROUND: Saccharin is a common artificial sweetener and a bona fide ligand for sweet taste receptors (STR). STR can regulate insulin secretion in beta cells, so we investigated whether saccharin can stimulate insulin secretion dependent on STR and the activation of phospholipase C (PLC) signaling. METHODS: We performed in vivo and in vitro approaches in mice and cells with loss-of-function of STR signaling and specifically assessed the involvement of a PLC signaling cascade using real-time biosensors and calcium imaging. RESULTS: We found that the ingestion of a physiological amount of saccharin can potentiate insulin secretion dependent on STR. Similar to natural sweeteners, saccharin triggers the activation of the PLC signaling cascade, leading to calcium influx and the vesicular exocytosis of insulin. The effects of saccharin also partially require transient receptor potential cation channel M5 (TRPM5) activity. CONCLUSIONS: Saccharin ingestion may transiently potentiate insulin secretion through the activation of the canonical STR signaling pathway. These physiological effects provide a framework for understanding the potential health impact of saccharin use and the contribution of STR in peripheral tissues.

5.
Mol Metab ; 54: 101343, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34583010

RESUMO

Regulation of organismal homeostasis in response to nutrient availability is a vital physiological process that involves inter-organ communication. Understanding the mechanisms controlling systemic cross-talk for the maintenance of metabolic health is critical to counteract diet-induced obesity. Here, we show that cardiac-derived transforming growth factor beta 1 (TGF-ß1) protects against weight gain and glucose intolerance in mice subjected to high-fat diet. Secretion of TGF-ß1 by cardiomyocytes correlates with the bioavailability of this factor in circulation. TGF-ß1 prevents adipose tissue inflammation independent of body mass and glucose metabolism phenotypes, indicating protection from adipocyte dysfunction-driven immune cell recruitment. TGF-ß1 alters the gene expression programs in white adipocytes, favoring their fatty acid oxidation and consequently increasing their mitochondrial oxygen consumption rates. Ultimately, subcutaneous and visceral white adipose tissue from cadiac-specific TGF-ß1 transgenic mice fail to undergo cellular hypertrophy, leading to reduced overall adiposity during high-fat feeding. Thus, TGF-ß1 is a critical mediator of heart-fat communication for the regulation of systemic metabolism.


Assuntos
Tecido Adiposo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Miócitos Cardíacos/metabolismo , Obesidade/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Feminino , Intolerância à Glucose , Masculino , Camundongos , Camundongos Transgênicos , Aumento de Peso
6.
Mol Metab ; 54: 101339, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34509698

RESUMO

OBJECTIVE: Sweet taste receptors (STR) are expressed in the gut and other extra-oral tissues, suggesting that STR-mediated nutrient sensing may contribute to human physiology beyond taste. A common variant (Ile191Val) in the TAS1R2 gene of STR is associated with nutritional and metabolic outcomes independent of changes in taste perception. It is unclear whether this polymorphism directly alters STR function and how it may contribute to metabolic regulation. METHODS: We implemented a combination of in vitro biochemical approaches to decipher the effects of TAS1R2 polymorphism on STR function. Then, as proof-of-concept, we assessed its effects on glucose homeostasis in apparently healthy lean participants. RESULTS: The Ile191Val variant causes a partial loss of function of TAS1R2 through reduced receptor availability in the plasma membrane. Val minor allele carriers have reduced glucose excursions during an OGTT, mirroring effects previously seen in mice with genetic loss of function of TAS1R2. These effects were not due to differences in beta-cell function or insulin sensitivity. CONCLUSIONS: Our pilot studies on a common TAS1R2 polymorphism suggest that STR sensory function in peripheral tissues, such as the intestine, may contribute to the regulation of metabolic control in humans.


Assuntos
Glucose/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Paladar/genética , Adulto , Feminino , Células HEK293 , Humanos , Masculino
7.
Microbiome ; 9(1): 11, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33431052

RESUMO

BACKGROUND: Non-caloric artificial sweeteners (NCAS) are widely used as a substitute for dietary sugars to control body weight or glycemia. Paradoxically, some interventional studies in humans and rodents have shown unfavorable changes in glucose homeostasis in response to NCAS consumption. The causative mechanisms are largely unknown, but adverse changes in gut microbiota have been proposed to mediate these effects. These findings have raised concerns about NCAS safety and called into question their broad use, but further physiological and dietary considerations must be first addressed before these results are generalized. We also reasoned that, since NCAS are bona fide ligands for sweet taste receptors (STRs) expressed in the intestine, some metabolic effects associated with NCAS use could be attributed to a common mechanism involving the host. RESULTS: We conducted a double-blind, placebo-controlled, parallel arm study exploring the effects of pure saccharin compound on gut microbiota and glucose tolerance in healthy men and women. Participants were randomized to placebo, saccharin, lactisole (STR inhibitor), or saccharin with lactisole administered in capsules twice daily to achieve the maximum acceptable daily intake for 2 weeks. In parallel, we performed a 10-week study administering pure saccharin at a high dose in the drinking water of chow-fed mice with genetic ablation of STRs (T1R2-KO) and wild-type (WT) littermate controls. In humans and mice, none of the interventions affected glucose or hormonal responses to an oral glucose tolerance test (OGTT) or glucose absorption in mice. Similarly, pure saccharin supplementation did not alter microbial diversity or composition at any taxonomic level in humans and mice alike. No treatment effects were also noted in readouts of microbial activity such as fecal metabolites or short-chain fatty acids (SCFA). However, compared to WT, T1R2-KO mice were protected from age-dependent increases in fecal SCFA and the development of glucose intolerance. CONCLUSIONS: Short-term saccharin consumption at maximum acceptable levels is not sufficient to alter gut microbiota or induce glucose intolerance in apparently healthy humans and mice. TRIAL REGISTRATION: Trial registration number NCT03032640 , registered on January 26, 2017. Video abstract.


Assuntos
Microbioma Gastrointestinal , Intolerância à Glucose , Voluntários Saudáveis , Sacarina/administração & dosagem , Sacarina/farmacologia , Adulto , Animais , Método Duplo-Cego , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Intolerância à Glucose/induzido quimicamente , Humanos , Masculino , Camundongos , Adulto Jovem
8.
Am J Clin Nutr ; 113(1): 232-245, 2021 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-33300030

RESUMO

In November 2019, the NIH held the "Sensory Nutrition and Disease" workshop to challenge multidisciplinary researchers working at the interface of sensory science, food science, psychology, neuroscience, nutrition, and health sciences to explore how chemosensation influences dietary choice and health. This report summarizes deliberations of the workshop, as well as follow-up discussion in the wake of the current pandemic. Three topics were addressed: A) the need to optimize human chemosensory testing and assessment, B) the plasticity of chemosensory systems, and C) the interplay of chemosensory signals, cognitive signals, dietary intake, and metabolism. Several ways to advance sensory nutrition research emerged from the workshop: 1) refining methods to measure chemosensation in large cohort studies and validating measures that reflect perception of complex chemosensations relevant to dietary choice; 2) characterizing interindividual differences in chemosensory function and how they affect ingestive behaviors, health, and disease risk; 3) defining circuit-level organization and function that link and interact with gustatory, olfactory, homeostatic, visceral, and cognitive systems; and 4) discovering new ligands for chemosensory receptors (e.g., those produced by the microbiome) and cataloging cell types expressing these receptors. Several of these priorities were made more urgent by the current pandemic because infection with sudden acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease of 2019 has direct short- and perhaps long-term effects on flavor perception. There is increasing evidence of functional interactions between the chemosensory and nutritional sciences. Better characterization of this interface is expected to yield insights to promote health, mitigate disease risk, and guide nutrition policy.

9.
Mol Metab ; 17: 98-111, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201274

RESUMO

OBJECTIVE: Beyond the taste buds, sweet taste receptors (STRs; T1R2/T1R3) are also expressed on enteroendocrine cells, where they regulate gut peptide secretion but their regulatory function within the intestine is largely unknown. METHODS: Using T1R2-knock out (KO) mice we evaluated the role of STRs in the regulation of glucose absorption in vivo and in intact intestinal preparations ex vivo. RESULTS: STR signaling enhances the rate of intestinal glucose absorption specifically in response to the ingestion of a glucose-rich meal. These effects were mediated specifically by the regulation of GLUT2 transporter trafficking to the apical membrane of enterocytes. GLUT2 translocation and glucose transport was dependent and specific to glucagon-like peptide 2 (GLP-2) secretion and subsequent intestinal neuronal activation. Finally, high-sucrose feeding in wild-type mice induced rapid downregulation of STRs in the gut, leading to reduced glucose absorption. CONCLUSIONS: Our studies demonstrate that STRs have evolved to modulate glucose absorption via the regulation of its transport and to prevent the development of exacerbated hyperglycemia due to the ingestion of high levels of sugars.


Assuntos
Glucose/metabolismo , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Transporte Biológico , Metabolismo Energético , Células Enteroendócrinas/metabolismo , Feminino , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Absorção Intestinal/efeitos dos fármacos , Jejuno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/deficiência , Transdução de Sinais/efeitos dos fármacos , Paladar
10.
Am J Clin Nutr ; 105(4): 1001-1009, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28251932

RESUMO

Background: Glucose is a natural ligand for sweet taste receptors (STRs) that are expressed on the tongue and in the gastrointestinal tract. Whether STRs directly contribute to the regulation of glucose homeostasis in response to glucose ingestion is unclear.Objective: We sought to determine the metabolic effects of the pharmacologic inhibition of STRs in response to an oral glucose load in healthy lean participants.Design: Ten healthy lean participants with a body mass index (in kg/m2) of 22.4 ± 0.8 were subjected to an oral-glucose-tolerance test (OGTT) on 4 separate days with the use of a randomized crossover design. Ten minutes before the 75-g OGTT, participants consumed a preload solution of either 300 parts per million (ppm) saccharin or water with or without the addition of 500 ppm lactisole, a human-specific inhibitor of STRs. When present, lactisole was included in both the preload and OGTT solutions. We assessed plasma responses of glucose, insulin, C-peptide, glucagon, glucagon-like peptides 1 and 2, gastric inhibitory peptide, acetaminophen, and 3-O-methylglucose. With the use of mathematical modeling, we estimated gastric emptying, glucose absorption, ß-cell function, insulin sensitivity and clearance, and the portal insulin:glucagon ratio.Results: The addition of lactisole to the OGTT caused increases in the plasma responses of insulin (P = 0.012), C-peptide (P = 0.004), and the insulin secretory rate (P = 0.020) compared with the control OGTT. The addition of lactisole also caused a slight reduction in the insulin sensitivity index independent of prior saccharin consumption (P < 0.025). The ingestion of saccharin before the OGTT did not alter any of the measured variables but eliminated the effects of lactisole on the OGTT.Conclusion: The pharmacologic inhibition of STRs in the gastrointestinal tract alters insulin responses during an oral glucose challenge in lean healthy participants. This trial was registered at clinicaltrials.gov as NCT02835859.


Assuntos
Derivados de Benzeno/farmacologia , Trato Gastrointestinal/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Paladar/fisiologia , Adulto , Glicemia/metabolismo , Índice de Massa Corporal , Proteína C-Reativa/metabolismo , Células Quimiorreceptoras , Estudos Cross-Over , Feminino , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Resistência à Insulina , Secreção de Insulina , Masculino , Receptores de Superfície Celular/fisiologia , Valores de Referência , Sacarina
11.
Am J Physiol Endocrinol Metab ; 310(8): E688-E698, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26884387

RESUMO

Sweet taste receptors (STRs) on the tongue mediate gustatory sweet sensing, but their expression in the gut, pancreas, and adipose tissue suggests a physiological contribution to whole body nutrient sensing and metabolism. However, little is known about the function and contribution of these sugar sensors during metabolic stress induced by overnutrition and subsequent obesity. Here, we investigated the effects of high-fat/low-carbohydrate (HF/LC) diet on glucose homeostasis and energy balance in mice with global disruption of the sweet taste receptor protein T1R2. We assessed body composition, energy balance, glucose homeostasis, and tissue-specific nutrient metabolism in T1R2 knockout (T1R2-KO) mice fed a HF/LC diet for 12 wk. HF/LC diet-fed T1R2-KO mice gained a similar amount of body mass as did WT mice, but had reduced fat mass and increased lean mass relative to WT mice. T1R2-KO mice were also hyperphagic and hyperactive. Ablation of the T1R2 sugar sensor protected mice from HF/LC diet-induced hyperinsulinemia and altered substrate utilization, including increased rates of glucose oxidation and decreased liver triglyceride (TG) accumulation, despite normal intestinal fat absorption. Finally, STRs (T1r2/T1r3) were upregulated in the adipose tissue of WT mice in response to HF/LC diet, and their expression positively correlated with fat mass and glucose intolerance. The chemosensory receptor T1R2, plays an important role in glucose homeostasis during diet-induced obesity through the regulation of yet to be identified molecular mechanisms that alter energy disposal and utilization in peripheral tissues.


Assuntos
Glicemia/metabolismo , Composição Corporal/genética , Dieta com Restrição de Carboidratos , Dieta Hiperlipídica , Metabolismo Energético/genética , Intolerância à Glucose/genética , Obesidade/genética , Receptores Acoplados a Proteínas G/genética , Tecido Adiposo/metabolismo , Aminoácidos , Animais , Peso Corporal/genética , Cromo , Intolerância à Glucose/metabolismo , Homeostase , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Ácidos Nicotínicos , Obesidade/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 3 Toll-Like/metabolismo , Triglicerídeos/metabolismo , Regulação para Cima
12.
Endocrinology ; 155(6): 2112-21, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24712876

RESUMO

ß-Cells rapidly secrete insulin in response to acute increases in plasma glucose but, upon further continuous exposure to glucose, insulin secretion progressively decreases. Although the mechanisms are unclear, this mode of regulation suggests the presence of a time-dependent glucosensory system that temporarily attenuates insulin secretion. Interestingly, early-stage ß-cell dysfunction is often characterized by basal (ie, fasting) insulin hypersecretion, suggesting a disruption of these related mechanisms. Because sweet taste receptors (STRs) on ß-cells are implicated in the regulation of insulin secretion and glucose is a bona fide STR ligand, we tested whether STRs mediate this sensory mechanism and participate in the regulation of basal insulin secretion. We used mice lacking STR signaling (T1R2(-/-) knockout) and pharmacologic inhibition of STRs in human islets. Mouse and human islets deprived of STR signaling hypersecrete insulin at short-term fasting glucose concentrations. Accordingly, 5-hour fasted T1R2(-/-) mice have increased plasma insulin and lower glucose. Exposure of isolated wild-type islets to elevated glucose levels reduced STR expression, whereas islets from diabetic (db/db) or diet-induced obese mouse models show similar down-regulation. This transcriptional reprogramming in response to hyperglycemia correlates with reduced STR function in these mouse models, leading to insulin hypersecretion. These findings reveal a novel mechanism by which insulin secretion is physiologically regulated by STRs and also suggest that, during the development of diabetes, STR function is compromised by hyperglycemia leading to hyperinsulinemia. These observations further suggest that STRs might be a promising therapeutic target to prevent and treat type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Humanos , Secreção de Insulina , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/genética
13.
Proc Natl Acad Sci U S A ; 109(8): E524-32, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22315413

RESUMO

Postprandial insulin release is regulated by glucose, but other circulating nutrients may target beta cells and potentiate glucose-stimulated insulin secretion via distinct signaling pathways. We demonstrate that fructose activates sweet taste receptors (TRs) on beta cells and synergizes with glucose to amplify insulin release in human and mouse islets. Genetic ablation of the sweet TR protein T1R2 obliterates fructose-induced insulin release and its potentiating effects on glucose-stimulated insulin secretion in vitro and in vivo. TR signaling in beta cells is triggered, at least in part, in parallel with the glucose metabolic pathway and leads to increases in intracellular calcium that are dependent on the activation of phospholipase C (PLC) and transient receptor potential cation channel, subfamily M, member 5 (TRPM5). Our results unveil a pathway for the regulation of insulin release by postprandial nutrients that involves beta cell sweet TR signaling.


Assuntos
Frutose/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Paladar/efeitos dos fármacos , Animais , Cálcio/metabolismo , Ativação Enzimática/efeitos dos fármacos , Deleção de Genes , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Camundongos , Período Pós-Prandial/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Fosfolipases Tipo C/metabolismo
14.
Hum Mol Genet ; 21(5): 963-77, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22045699

RESUMO

Endoplasmic reticulum (ER) stress has been implicated as an initiator or contributing factor in neurodegenerative diseases. The mechanisms that lead to ER stress and whereby ER stress contributes to the degenerative cascades remain unclear but their understanding is critical to devising effective therapies. Here we show that knockdown of Herp (Homocysteine-inducible ER stress protein), an ER stress-inducible protein with an ubiquitin-like (UBL) domain, aggravates ER stress-mediated cell death induced by mutant α-synuclein (αSyn) that causes an inherited form of Parkinson's disease (PD). Functionally, Herp plays a role in maintaining ER homeostasis by facilitating proteasome-mediated degradation of ER-resident Ca(2+) release channels. Deletion of the UBL domain or pharmacological inhibition of proteasomes abolishes the Herp-mediated stabilization of ER Ca(2+) homeostasis. Furthermore, knockdown or pharmacological inhibition of ER Ca(2+) release channels ameliorates ER stress, suggesting that impaired homeostatic regulation of Ca(2+) channels promotes a protracted ER stress with the consequent activation of ER stress-associated apoptotic pathways. Interestingly, sustained upregulation of ER stress markers and aberrant accumulation of ER Ca(2+) release channels were detected in transgenic mutant A53T-αSyn mice. Collectively, these data establish a causative link between impaired ER Ca(2+) homeostasis and chronic ER stress in the degenerative cascades induced by mutant αSyn and suggest that Herp is essential for the resolution of ER stress through maintenance of ER Ca(2+) homeostasis. Our findings suggest a therapeutic potential in PD for agents that increase Herp levels or its ER Ca(2+)-stabilizing action.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/fisiologia , Proteínas de Membrana/metabolismo , Estresse Fisiológico , alfa-Sinucleína/metabolismo , Animais , Canais de Cálcio/metabolismo , Morte Celular , Degradação Associada com o Retículo Endoplasmático , Células HEK293 , Homeostase , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/metabolismo , Células PC12 , Interferência de RNA , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , alfa-Sinucleína/genética
15.
Cancer Res ; 70(1): 418-27, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20028870

RESUMO

Glioblastoma multiforme (GBM) is the most frequent and incurable type of brain tumor of adults. Hypoxia has been shown to direct GBM toward a more aggressive and malignant state. Here we show that hypoxia increases Notch1 activation, which in turn induces the expression of transient receptor potential 6 (TRPC6) in primary samples and cell lines derived from GBM. TRPC6 is required for the development of the aggressive phenotype because knockdown of TRPC6 expression inhibits glioma growth, invasion, and angiogenesis. Functionally, TRPC6 causes a sustained elevation of intracellular calcium that is coupled to the activation of the calcineurin-nuclear factor of activated T-cell (NFAT) pathway. Pharmacologic inhibition of the calcineurin-NFAT pathway substantially reduces the development of the malignant GBM phenotypes under hypoxia. Clinically, expression of TRPC6 was elevated in GBM specimens in comparison with normal tissues. Collectively, our studies indicate that TRPC6 is a key mediator of tumor growth of GBM in vitro and in vivo and that TRPC6 may be a promising therapeutic target in the treatment of human GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Invasividade Neoplásica/patologia , Receptor Notch1/metabolismo , Canais de Cátion TRPC/metabolismo , Adulto , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Hipóxia Celular/fisiologia , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Invasividade Neoplásica/genética , RNA Interferente Pequeno , Receptor Notch1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6
16.
J Biol Chem ; 285(9): 6811-25, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20038578

RESUMO

The Notch signaling pathway plays an essential role in the regulation of cell specification by controlling differentiation, proliferation, and apoptosis. Numb is an intrinsic regulator of the Notch pathway and exists in four alternative splice variants that differ in the length of their phosphotyrosine-binding domain (PTB) and proline-rich region domains. The physiological relevance of the existence of the Numb splice variants and their exact regulation are still poorly understood. We previously reported that Numb switches from isoforms containing the insertion in PTB to isoforms lacking this insertion in neuronal cells subjected to trophic factor withdrawal (TFW). The functional relevance of the TFW-induced switch in Numb isoforms is not known. Here we provide evidence that the TFW-induced switch in Numb isoforms regulates Notch signaling strength and Notch target gene expression. PC12 cells stably overexpressing Numb isoforms lacking the PTB insertion exhibited higher basal Notch activity and Notch-dependent transcription of the transient receptor potential channel 6 (TRPC6) when compared with those overexpressing Numb isoforms with the PTB insertion. The differential regulation of TRPC6 expression is correlated with perturbed calcium signaling and increased neuronal vulnerability to TFW-induced death. Pharmacological inhibition of the Notch pathway or knockdown of TRPC6 function ameliorates the adverse effects caused by the TFW-induced switch in Numb isoforms. Taken together, our results indicate that Notch and Numb interaction may influence the sensitivity of neuronal cells to injurious stimuli by modulating calcium-dependent apoptotic signaling cascades.


Assuntos
Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores Notch/metabolismo , Canais de Cátion TRPC/genética , Animais , Sinalização do Cálcio , Morte Celular , Humanos , Neurônios/metabolismo , Células PC12 , Isoformas de Proteínas , Ratos , Transdução de Sinais , Estresse Fisiológico , Regulação para Cima/genética
17.
J Biol Chem ; 284(27): 18323-33, 2009 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-19447887

RESUMO

The endoplasmic reticulum (ER) is a key organelle regulating intracellular Ca(2+) homeostasis. Oxidants and mitochondria-derived free radicals can target ER-based Ca(2+) regulatory proteins and cause uncontrolled Ca(2+) release that may contribute to protracted ER stress and apoptosis. Several ER stress proteins have been suggested to counteract the deregulation of ER Ca(2+) homeostasis and ER stress. Here we showed that knockdown of Herp, an ubiquitin-like domain containing ER stress protein, renders PC12 and MN9D cells vulnerable to 1-methyl-4-phenylpyridinium-induced cytotoxic cell death by a mechanism involving up-regulation of CHOP expression and ER Ca(2+) depletion. Conversely, Herp overexpression confers protection by blocking 1-methyl-4-phenylpyridinium-induced CHOP up-regulation, ER Ca(2+) store depletion, and mitochondrial Ca(2+) accumulation in a manner dependent on a functional ubiquitin-proteasomal protein degradation pathway. Deletion of the ubiquitin-like domain of Herp or treatment with a proteasomal inhibitor abolished the central function of Herp in ER Ca(2+) homeostasis. Thus, elucidating the underlying molecular mechanism(s) whereby Herp counteracts Ca(2+) disturbances will provide insights into the molecular cascade of cell death in dopaminergic neurons and may uncover novel therapeutic strategies to prevent and ameliorate Parkinson disease progression.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cálcio/metabolismo , Intoxicação por MPTP/fisiopatologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neurônios/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Retículo Endoplasmático/metabolismo , Homeostase/fisiologia , Humanos , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/patologia , Proteínas de Membrana/química , Camundongos , Neurônios/citologia , Células PC12 , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno , Ratos , Estresse Fisiológico/fisiologia , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Transfecção , Ubiquitina/metabolismo
18.
J Biol Chem ; 283(37): 25492-25502, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18599481

RESUMO

Central to the pathogenesis of Alzheimer disease is the aberrant processing of the amyloid precursor protein (APP) to generate amyloid beta-peptide (Abeta), the principle component of amyloid plaques. The cell fate determinant Numb is a phosphotyrosine binding domain (PTB)-containing endocytic adapter protein that interacts with the carboxyl-terminal domain of APP. The physiological relevance of this interaction is unknown. Mammals produce four alternatively spliced variants of Numb that differ in the length of their PTB and proline-rich region. In the current study, we determined the influence of the four human Numb isoforms on the intracellular trafficking and processing of APP. Stable expression of Numb isoforms that differ in the PTB but not in the proline-rich region results in marked differences in the sorting of APP to the recycling and degradative pathways. Neural cells expressing Numb isoforms that lack the insert in the PTB (short PTB (SPTB)) exhibited marked accumulation of APP in Rab5A-labeled early endosomal and recycling compartments, whereas those expressing isoforms with the insertion in the PTB (long PTB (LPTB)) exhibited reduced amounts of cellular APP and its proteolytic derivatives relative to parental control cells. Neither the activities of the beta- and gamma-secretases nor the expression of APP mRNA were significantly different in the stably transfected cells, suggesting that the differential effects of the Numb proteins on APP metabolism is likely to be secondary to altered APP trafficking. In addition, the expression of SPTB-Numb increases at the expense of LPTB-Numb in neuronal cultures subjected to stress, suggesting a role for Numb in stress-induced Abeta production. Taken together, these results suggest distinct roles for the human Numb isoforms in APP metabolism and may provide a novel potential link between altered Numb isoform expression and increased Abeta generation.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/química , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Endocitose , Humanos , Microscopia de Fluorescência , Modelos Biológicos , Neurônios/metabolismo , Células PC12 , Nexinas de Proteases , Isoformas de Proteínas , Ratos , Receptores de Superfície Celular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
19.
Eur J Appl Physiol ; 101(5): 621-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17701049

RESUMO

Apolipoprotein B (apoB) concentration and age are independently associated with an increased risk for cardiovascular disease. Age is also associated with increased apoB concentration. The purpose of this study was to determine the effects of exercise on apoB and examine the association between age and lipoproteins. Forty-one sedentary individuals exercised for 6 months, four times/week for 40 min between 60 and 85% of their maximal heart rate. Lipids were determined three times: before training, 24 and 72 h after the last training session. Exercise did not alter apoB (1.2+/-0.05 g/l vs. 1.2+/-0.05 g/l; P>0.05), or other lipids or lipoproteins. When participants were sequestered by baseline low density lipoprotein cholesterol (LDLc), total cholesterol (TC) was decreased at 24 h post (6.3+/-0.2 mmol/l vs. 6.0+/-0.2 mmol/l, P<0.05) and LDLc after 24 and 48 h post (4.3+/-0.1 mg/dl vs. 3.9+/-0.1 and 4.1+/-0.2 mg/dl, P<0.05) in the high LDLc group. In the low LDLc group both TC (4.4+/-0.2 mmol/l vs. 4.6+/-0.2 and 4.6+/-0.2 mmol/l, P>0.05) and LDLc (2.6+/-0.1 mmol/l vs. 2.8+/-0.1 and 2.8+/-0.2 mmol/l, P<0.05) were elevated at 24 h and remained elevated at 72 h post compared to baseline. Age does not affect apoB or lipoproteins in response to exercise. Individuals with high baseline LDLc experienced acute reduction in TC and LDLc produced by each exercise session.


Assuntos
Envelhecimento/fisiologia , Apolipoproteínas B/sangue , LDL-Colesterol/sangue , Colesterol/sangue , Exercício Físico/fisiologia , Lipoproteínas/sangue , Aptidão Física/fisiologia , Adolescente , Adulto , Algoritmos , Limiar Anaeróbio/fisiologia , Estatura/fisiologia , Índice de Massa Corporal , Peso Corporal/fisiologia , Dieta , Feminino , Humanos , Lipase/sangue , Lipídeos/sangue , Lipase Lipoproteica/sangue , Fígado/enzimologia , Masculino , Pessoa de Meia-Idade
20.
Clin J Sport Med ; 17(1): 49-51, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17304006

RESUMO

OBJECTIVE: The purpose of the study was to determine whether exercise-induced increases in energy expenditure (EE) alter circulating leptin levels in obese individuals. DESIGN: Participants were randomized to an exercise intervention group (n = 8) or nonexercising control (n = 7). SETTING: All data were collected on an outpatient basis at the exercise physiology laboratory at the University of Central Florida. PATIENTS: Fifteen healthy obese males (24.9 +/- 1.4 years old, body mass index 33.4 +/- 0.7 kg . m). INTERVENTIONS: Members of the intervention group underwent a single exercise session of moderate intensity (58.4 +/- 1.3% of VO2max) for 60 minutes. MAIN OUTCOME MEASUREMENTS: Postexercise, 24 hour postexercise, and 48 hour postexercise levels of leptin, insulin, and ghrelin. RESULTS: The exercise session elicited an EE of 567 +/- 25 Kcal. No significant main effect or time-by-group interactions for leptin or ghrelin were observed immediately after the exercise bout or in the days following the intervention. CONCLUSIONS: These preliminary data suggest that a bout of acute exercise of moderate intensity and duration does not affect leptin concentration. It is possible that a higher level of EE is required to elicit substantial changes.


Assuntos
Metabolismo Energético , Exercício Físico/fisiologia , Leptina/fisiologia , Atividade Motora/fisiologia , Obesidade/fisiopatologia , Adulto , Grelina , Humanos , Leptina/sangue , Masculino , Hormônios Peptídicos/sangue , Estudos Prospectivos , Fatores Sexuais , Inquéritos e Questionários , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...