Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Endocr J ; 69(5): 487-493, 2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-34803124

RESUMO

T1R3 is a class C G protein-coupled receptor family member that forms heterodimeric umami and sweet taste receptors with T1R1 and T1R2, respectively, in the taste cells of taste buds. T1R3 is expressed in 3T3-L1 cells in homomeric form and negatively regulates adipogenesis in a Gαs-dependent but cAMP-independent manner. Although T1R3 expression is markedly upregulated during adipogenesis, its physiological role in mature adipocytes remains obscure. Here, we show that stimulation of T1R3 with sucralose or saccharin induces microtubule disassembly in differentiated 3T3-L1 adipocytes. The effect was reproduced by treatment with cholera toxin or isoproterenol but not with forskolin. Treatment with sucralose or saccharin for 3 h inhibited insulin-stimulated glucose uptake by 32% and 45% in differentiated adipocytes, respectively, similar to the inhibitory effect of nocodazole (by 33%). Isoproterenol treatment inhibited insulin-stimulated glucose transport by 26%, whereas sucralose did not affect the intrinsic activity of the glucose transporter, indicating that it inhibited insulin-induced GLUT4 translocation to the plasma membrane. Immunostaining analysis showed that insulin-stimulated GLUT4 accumulation on the plasma membrane was abrogated in sucralose-treated cells, in association with depolymerization of microtubules. Sucralose-mediated inhibition of GLUT4 translocation was reversed by the overexpression of dominant-negative Gαs (Gαs-G226A) or knockdown of Gαs. Additionally, membrane fractionation analysis showed that sucralose treatment reduced GLUT4 levels in the plasma membrane fraction from insulin-stimulated adipocytes. We have identified a novel non-gustatory role for homomeric T1R3 in adipocytes, and activation of the T1R3 receptor negatively regulates insulin action of glucose transport via Gαs-dependent microtubule disassembly.


Assuntos
Papilas Gustativas , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Isoproterenol/metabolismo , Isoproterenol/farmacologia , Camundongos , Microtúbulos/metabolismo , Sacarina/metabolismo , Paladar , Papilas Gustativas/metabolismo
2.
J Endocrinol ; 245(1): 65-78, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31990671

RESUMO

Despite extensive investigation, the mechanisms underlying adipogenesis are not fully understood. We previously identified proliferative cells in adipose tissue expressing adipocyte-specific genes, which were named small proliferative adipocytes (SPA). In this study, we investigated the characteristics and roles of SPA in adipose tissue. Epididymal and inguinal fat was digested by collagenase, and then SPA were separated by centrifugation from stromal vascular cells (SVC) and mature white adipocytes. To clarify the feature of gene expression in SPA, microarray and real-time PCR were performed. The expression of adipocyte-specific genes and several neuronal genes was increased in the order of SVC < SPA < mature white adipocytes. In addition, proliferin was detected only in SPA. SPA differentiated more effectively into lipid-laden cells than SVC. Moreover, differentiated SPA expressed uncoupling protein 1 and mitochondria-related genes more than differentiated SVC. Treatment of SPA with pioglitazone and CL316243, a specific ß3-adrenergic receptor agonist, differentiated SPA into beige-like cells. Therefore, SPA are able to differentiate into beige cells. SPA isolated from epididymal fat (epididymal SPA), but not SPA from inguinal fat (inguinal SPA), expressed a marker of visceral adipocyte precursor, WT1. However, no significant differences were detected in the expression levels of adipocyte-specific genes or neuronal genes between epididymal and inguinal SPA. The ability to differentiate into lipid-laden cells in epididymal SPA was a little superior to that in inguinal SPA, whereas the ability to differentiate into beige-like cells was greater in inguinal SPA than epididymal SPA. In conclusion, SPA may be progenitors of beige cells.


Assuntos
Adipócitos Bege/metabolismo , Adipócitos Brancos/metabolismo , Adipócitos/metabolismo , Perfilação da Expressão Gênica/métodos , Células-Tronco/metabolismo , Adipócitos/citologia , Adipócitos Bege/citologia , Adipócitos Brancos/citologia , Adipogenia/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Células-Tronco/citologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
3.
BMC Pulm Med ; 19(1): 70, 2019 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-30914062

RESUMO

BACKGROUND: Defective phagocytosis in alveolar macrophages is associated with chronic obstructive pulmonary disease (COPD). Transient receptor potential cation channel subfamily V member 2 (TRPV2), a type of nonselective cation channel pertinent to diverse physiological functions, regulates macrophage phagocytosis. However, the role of TRPV2 in COPD remains poorly understood. Here, we explored the role of TRPV2 in the development of COPD. METHODS: Macrophage TRPV2 expression and phagocytosis function were measured in MH-S cells (a murine alveolar macrophage cell line) and a cigarette smoke exposure mouse model. RESULTS: TRPV2 expression and phagocytosis function were reduced when MH-S cells were exposed to cigarette smoke extract (CSE). TRPV2 knockdown by siRNA decreased phagocytosis in MH-S cells. Consistently, TRPV2 expression was reduced in alveolar macrophages prepared from bronchoalveolar lavage samples of mice which were exposed to cigarette smoke for 2 months. In addition, the alveolar space was progressively enlarged during development in TRPV2 knockout (TRPV2KO) mice. Moreover, exposure to cigarette smoke for 2 months significantly induced alveolar space enlargement in TRPV2KO mice, but not in wild-type (WT) mice. The phagocytic function of alveolar macrophages from TRPV2KO mice was reduced, compared with macrophages from WT mice. CONCLUSIONS: TRPV2 expression is profoundly downregulated in alveolar macrophages at early time points of cigarette smoke exposure. Reduced TRPV2-mediated phagocytic function renders the lung susceptible to cigarette smoke-induced alveolar space enlargement. TRPV2 may provide a therapeutic target for COPD induced by cigarette smoke.


Assuntos
Canais de Cálcio/metabolismo , Pulmão/patologia , Macrófagos Alveolares/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Canais de Cálcio/genética , Linhagem Celular , Células Cultivadas , Fumar Cigarros , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Canais de Cátion TRPV/genética
4.
Pflugers Arch ; 471(4): 647-654, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30552496

RESUMO

Over recent years, the presence of the sweet taste receptor TIR3 in rodent and human insulin-producing pancreatic islet ß-cells was documented. The activation of this receptor by sweet-tasting sucralose mimics several biochemical and functional effects of D-glucose in the ß-cells. The present study extends this analogy to the bioelectrical response of ß-cells. In this respect, sucralose was inefficient in the absence of D-glucose, but induced on occasion electrical activity in mouse ß-cells exposed to low non-stimulatory concentrations of the hexose and potentiated, in a concentration-related manner, the response to stimulatory concentrations of D-glucose. These data indicate that sucralose, acting as an agonist of the TIR3 receptor, exerts an excitatory effect upon pancreatic ß-cell bioelectrical activity.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Papilas Gustativas/fisiologia , Paladar/fisiologia , Animais , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Camundongos , Papilas Gustativas/metabolismo
5.
Hum Cell ; 31(2): 95-101, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29249016

RESUMO

Increasing metabolic syndromes including type-2 diabetes mellitus, obesity, and steatohepatitis are serious problems in most countries in the world. Neurodegenerative diseases such as Alzheimer, Parkinson's, and Huntington's diseases are increasing in many countries. However, therapy for these diseases is not sufficient yet. Thus, effective chemotherapy for these diseases is being expected. Conophylline is an alkaloid isolated from the leaves of Ervatamia microphylla and related plants. It was found to induce beta-cell differentiation in the precursor pancreatic cells. Oral administration of this compound ameliorated type-2 diabetes mellitus model in mice and rats. Later, fibrosis of the pancreatic islets was found to be greatly reduced by conophylline in the pancreatic islets. It also inhibited chemically induced liver cirrhosis. Further study indicated that conophylline inhibited non-alcoholic steatohepatitis in the model mice. On the one hand, loss of autophagy often causes protein aggregation to give neural cell death. Conophylline was found to activate autophagy in cultured neural cells. Activation of autophagy ameliorated cellular models of Parkinson's and Huntington's diseases. Thus, conophylline is likely to be useful for the development of chemotherapy for metabolic and neurodegenerative diseases.


Assuntos
Síndrome Metabólica/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fitoterapia , Alcaloides de Vinca/farmacologia , Alcaloides de Vinca/uso terapêutico , Animais , Autofagia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Fibrose , Humanos , Ilhotas Pancreáticas/patologia , Camundongos , Terapia de Alvo Molecular , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Folhas de Planta/química , Tabernaemontana/química , Alcaloides de Vinca/isolamento & purificação
6.
Diabetes Obes Metab ; 19 Suppl 1: 54-62, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28880472

RESUMO

Glucose is a primary stimulator of insulin secretion. It has been thought that glucose exerts its effect by a mechanism solely dependent on glucose metabolism. We show here that glucose induces rapid Ca2+ and cyclic AMP signals in ß-cells. These rapid signals are independent of glucose-metabolism and are reproduced by non-metabolizable glucose analogues. These results led us to postulate that glucose activates a cell-surface receptor, namely the glucose-sensing receptor. Rapid signals induced by glucose are blocked by inhibition of a sweet taste receptor subunit T1R3 and a calcium-sensing receptor subunit CaSR. In accordance with these observations, T1R3 and CaSR form a heterodimer. In addition, a heterodimer of T1R3 and CaSR is activated by glucose. These results suggest that a heterodimer of T1R3 and CaSR is a major component of the glucose-sensing receptor. When the glucose-sensing receptor is blocked, glucose-induced insulin secretion is inhibited. Also, ATP production is significantly attenuated by the inhibition of the receptor. Conversely, stimulation of the glucose-sensing receptor by either artificial sweeteners or non-metabolizable glucose analogue increases ATP. Hence, the glucose-sensing receptor signals promote glucose metabolism. Collectively, glucose activates the cell-surface glucose-sensing receptor and promotes its own metabolism. Glucose then enters the cells and is metabolized through already activated metabolic pathways. The glucose-sensing receptor is a key molecule regulating the action of glucose in ß-cells.


Assuntos
Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Modelos Biológicos , Receptores de Superfície Celular/agonistas , Animais , Sinalização do Cálcio , AMP Cíclico/metabolismo , Dimerização , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Multimerização Proteica , Receptores de Detecção de Cálcio/agonistas , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sistemas do Segundo Mensageiro
7.
Eur J Pharmacol ; 814: 130-137, 2017 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-28823924

RESUMO

GPBA is a G protein-coupled receptor that is activated by bile acids. Because activation of GPBA leads to increased cAMP levels and secretion of incretins and insulin, GPBA has been proposed as a promising drug target for the treatment of metabolic syndrome. Previously, we have developed a ligand-screening system to identify novel agonists of GPBA by means of a fusion protein of GPBA with G protein stimulatory α subunit (Gsα) and by a [35S]GTPγS-binding assay. To express the GPBA-Gsα fusion protein, transgenic silkworms were employed in this study, and cell membrane fractions were prepared from their fat body or pupae. We applied them to the screening of a chemical library that contains 10,625 compounds from the RIKEN Natural Products Depository (NPDepo). Eventually, a unique partial agonist, GUM2, was successfully identified. Our results indicated that the GPCR-Gα fusion proteins were beneficial for ligand identification and that the transgenic silkworms were useful for large-scale production of GPCRs. In HEK293 cells transiently expressing GPBA, GUM2 showed 50% effective concentration (EC50) of 3.5 ± 2.4µM and induced GPBA internalization as effectively as did an endogenous agonist, TLC. We also confirmed that GUM2 stimulates insulin secretion in MIN6 cells. Moreover, a single 2mg/kg dose of GUM2 significantly reduced blood glucose levels in mice during an intraperitoneal glucose tolerance test even though GUM2 is only a partial agonist with a low intrinsic activity. We concluded that GUM2 is a good candidate for research on GPBA signaling under physiological conditions and for the development of GPBA-targeting therapeutic compounds.


Assuntos
Produtos Biológicos/farmacologia , Glicemia/metabolismo , Teste de Tolerância a Glucose , Receptores Acoplados a Proteínas G/agonistas , Animais , Células HEK293 , Humanos , Insulina/metabolismo , Secreção de Insulina , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Larva/metabolismo , Camundongos , Pupa/metabolismo
9.
PLoS One ; 12(5): e0176841, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28472098

RESUMO

We previously reported that 3T3-L1 cells express a functional sweet taste receptor possibly as a T1R3 homomer that is coupled to Gs and negatively regulates adipogenesis by a Gαs-mediated but cAMP-independent mechanism. Here, we show that stimulation of this receptor with sucralose or saccharin induced disassembly of the microtubules in 3T3-L1 preadipocytes, which was attenuated by overexpression of the dominant-negative mutant of Gαs (Gαs-G226A). In contrast, overexpression of the constitutively active mutant of Gαs (Gαs-Q227L) as well as treatment with cholera toxin or isoproterenol but not with forskolin caused disassembly of the microtubules. Sweetener-induced microtubule disassembly was accompanied by activation of RhoA and Rho-associated kinase (ROCK). This was attenuated with by knockdown of GEF-H1, a microtubule-localized guanine nucleotide exchange factor for Rho GTPase. Furthermore, overexpression of the dominant-negative mutant of RhoA (RhoA-T19N) blocked sweetener-induced dephosphorylation of Akt and repression of PPARγ and C/EBPα in the early phase of adipogenic differentiation. These results suggest that the T1R3 homomeric sweet taste receptor negatively regulates adipogenesis through Gαs-mediated microtubule disassembly and consequent activation of the Rho/ROCK pathway.


Assuntos
Adipogenia/fisiologia , Cromograninas/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Microtúbulos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3-L1 , Animais , Transferência Ressonante de Energia de Fluorescência , Camundongos
10.
FASEB J ; 31(4): 1368-1381, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28007781

RESUMO

We have previously reported that transient receptor potential vanilloid 2 (TRPV2) can be activated by mechanical stimulation, which enhances axonal outgrowth in developing neurons; however, the molecular mechanisms that govern the contribution of TRPV2 activation to axonal outgrowth remain unclear. In the present study, we examined this mechanism by using PC12 cells as a neuronal model. Overexpression of TRPV2 enhanced axonal outgrowth in a mechanical stimulus-dependent manner. Accumulation of TRPV2 at the cell surface was 4-fold greater in the growth cone compared with the soma. In the growth cone, TRPV2 is not static, but dynamically accumulates (within ∼100 ms) to the site of mechanical stimulation. The dynamic and acute clustering of TRPV2 can enhance very weak mechanical stimuli via focal accumulation of TRPV2. Focal application of mechanical stimuli dramatically increased growth cone motility and caused actin reorganization via activation of TRPV2. We also found that TRPV2 physically interacts with actin and that changes in the actin cytoskeleton are required for its activation. Here, we demonstrated for the first time to our knowledge that TRPV2 clustering is induced by mechanical stimulation generated by axonal outgrowth and that TRPV2 activation is triggered by actin rearrangements that result from mechanical stimulation. Moreover, TRPV2 activation enhances growth cone motility and actin accumulation to promote axonal outgrowth. Sugio, S., Nagasawa, M., Kojima, I., Ishizaki, Y., Shibasaki, K. Transient receptor potential vanilloid 2 activation by focal mechanical stimulation requires interaction with the actin cytoskeleton and enhances growth cone motility.


Assuntos
Citoesqueleto de Actina/metabolismo , Cones de Crescimento/metabolismo , Crescimento Neuronal , Canais de Cátion TRPV/metabolismo , Animais , Mecanotransdução Celular , Células PC12 , Ligação Proteica , Ratos , Canais de Cátion TRPV/genética
11.
Front Neurosci ; 10: 502, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27877104

RESUMO

The hypothalamic feeding center plays an important role in energy homeostasis. In the feeding center, whole-body energy signals including hormones and nutrients are sensed, processed, and integrated. As a result, food intake and energy expenditure are regulated. Two types of glucose-sensing neurons exist in the hypothalamic arcuate nucleus (ARC): glucose-excited neurons and glucose-inhibited neurons. While some molecules are known to be related to glucose sensing in the hypothalamus, the mechanisms underlying glucose sensing in the hypothalamus are not fully understood. The sweet taste receptor is a heterodimer of taste type 1 receptor 2 (T1R2) and taste type 1 receptor 3 (T1R3) and senses sweet tastes. T1R2 and T1R3 are distributed in multiple organs including the tongue, pancreas, adipose tissue, and hypothalamus. However, the role of sweet taste receptors in the ARC remains to be clarified. To examine the role of sweet taste receptors in the ARC, cytosolic Ca2+ concentration ([Ca2+]i) in isolated single ARC neurons were measured using Fura-2 fluorescent imaging. An artificial sweetener, sucralose at 10-5-10-2 M dose dependently increased [Ca2+]i in 12-16% of ARC neurons. The sucralose-induced [Ca2+]i increase was suppressed by a sweet taste receptor inhibitor, gurmarin. The sucralose-induced [Ca2+]i increase was inhibited under an extracellular Ca2+-free condition and in the presence of an L-type Ca2+ channel blocker, nitrendipine. Sucralose-responding neurons were activated by high-concentration of glucose. This response to glucose was markedly suppressed by gurmarin. More than half of sucralose-responding neurons were activated by leptin but not ghrelin. Percentages of proopiomelanocortin (POMC) neurons among sucralose-responding neurons and sweet taste receptor expressing neurons were low, suggesting that majority of sucralose-responding neurons are non-POMC neurons. These data suggest that sweet taste receptor-mediated cellular activation mainly occurs on non-POMC leptin-responding neurons and contributes to glucose responding. Endogenous sweet molecules including glucose may regulate energy homeostasis through sweet taste receptors on glucose-and leptin-responsive neurons in the ARC.

12.
J Biol Chem ; 291(44): 23126-23135, 2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27613866

RESUMO

The calcium-sensing receptor (CaSR) is activated by various cations, cationic compounds, and amino acids. In the present study we investigated the effect of glucose on CaSR in HEK293 cells stably expressing human CaSR (HEK-CaSR cells). When glucose concentration in the buffer was raised from 3 to 25 mm, a rapid elevation of cytoplasmic Ca2+ concentration ([Ca2+]c) was observed. This elevation was immediate and transient and was followed by a sustained decrease in [Ca2+]c The effect of glucose was detected at a concentration of 4 mm and reached its maximum at 5 mm 3-O-Methylglucose, a non-metabolizable analogue of glucose, reproduced the effect of glucose. Sucrose also induced an elevation of [Ca2+]c in HEK-CaSR cells. Similarly, sucralose was nearly as effective as glucose in inducing elevation of [Ca2+]c Glucose was not able to increase [Ca2+]c in the absence of extracellular Ca2+ The effect of glucose on [Ca2+]c was inhibited by NPS-2143, an allosteric inhibitor of CaSR. In addition, NPS-2143 also inhibited the [Ca2+]c responses to sucralose and sucrose. Glucose as well as sucralose decreased cytoplasmic cAMP concentration in HEK-CaSR cells. The reduction of cAMP induced by glucose was blocked by pertussis toxin. Likewise, sucralose reduced [cAMP]c Finally, glucose increased [Ca2+]c in PT-r parathyroid cells and in Madin-Darby canine kidney cells, both of which express endogenous CaSR. These results indicate that glucose acts as a positive allosteric modulator of CaSR.


Assuntos
Glucose/metabolismo , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/metabolismo , Regulação Alostérica , Cálcio/metabolismo , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Glucose/análise , Células HEK293 , Humanos , Receptores de Detecção de Cálcio/genética
13.
Endocr J ; 63(8): 715-25, 2016 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-27250218

RESUMO

Sucralose is an artificial sweetener and activates the glucose-sensing receptor expressed in pancreatic ß-cells. Although sucralose does not enter ß-cells nor acts as a substrate for glucokinase, it induces a marked elevation of intracellular ATP ([ATP]c). The present study was conducted to identify the signaling pathway responsible for the elevation of [ATP]c induced by sucralose. Previous studies have shown that sucralose elevates cyclic AMP (cAMP), activates phospholipase C (PLC) and stimulates Ca(2+) entry by a Na(+)-dependent mechanism in MIN6 cells. The addition of forskolin induced a marked elevation of cAMP, whereas it did not affect [ATP]c. Carbachol, an activator of PLC, did not increase [ATP]c. In addition, activation of protein kinase C by dioctanoylglycerol did not affect [ATP]c. In contrast, nifedipine, an inhibitor of the voltage-dependent Ca(2+) channel, significantly reduced [ATP]c response to sucralose. Removal of extracellular Na(+) nearly completely blocked sucralose-induced elevation of [ATP]c. Stimulation of Na(+) entry by adding a Na(+) ionophore monensin elevated [ATP]c. The monensin-induced elevation of [ATP]c was only partially inhibited by nifedipine and loading of BAPTA, both of which completely abolished elevation of [Ca(2+)]c. These results suggest that Na(+) entry is critical for the sucralose-induced elevation of [ATP]c. Both calcium-dependent and -independent mechanisms are involved in the action of sucralose.


Assuntos
Trifosfato de Adenosina/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Células Secretoras de Insulina/efeitos dos fármacos , Sacarose/análogos & derivados , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Células Secretoras de Insulina/metabolismo , Camundongos , Nifedipino/farmacologia , Sacarose/farmacologia , Edulcorantes/farmacologia
14.
Endocrinology ; 157(5): 1839-51, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26943364

RESUMO

Sphingosine 1-phosphate (S1P) is known to regulate insulin resistance in hepatocytes, skeletal muscle cells, and pancreatic ß-cells. Among its 5 cognate receptors (S1pr1-S1pr5), S1P seems to counteract insulin signaling and confer insulin resistance via S1pr2 in these cells. S1P may also regulate insulin resistance in adipocytes, but the S1pr subtype(s) involved remains unknown. Here, we investigated systemic glucose/insulin tolerance and phenotypes of epididymal adipocytes in high-fat diet (HFD)-fed wild-type and S1pr2-deficient (S1pr2(-/-)) mice. Adult S1pr2(-/-) mice displayed smaller body/epididymal fat tissue weights, but the differences became negligible after 4 weeks with HFD. However, HFD-fed S1pr2(-/-) mice displayed better scores in glucose/insulin tolerance tests and had smaller epididymal adipocytes that expressed higher levels of proliferating cell nuclear antigen than wild-type mice. Next, proliferation/differentiation of 3T3-L1 and 3T3-F442A preadipocytes were examined in the presence of various S1pr antagonists: JTE-013 (S1pr2 antagonist), VPC-23019 (S1pr1/S1pr3 antagonist), and CYM-50358 (S1pr4 antagonist). S1P or JTE-013 treatment of 3T3-L1 preadipocytes potently activated their proliferation and Erk phosphorylation, whereas VPC-23019 inhibited both of these processes, and CYM-50358 had no effects. In contrast, S1P or JTE-013 treatment inhibited adipogenic differentiation of 3T3-F442A preadipocytes, whereas VPC-23019 activated it. The small interfering RNA knockdown of S1pr2 promoted proliferation and inhibited differentiation of 3T3-F442A preadipocytes, whereas that of S1pr1 acted oppositely. Moreover, oral JTE-013 administration improved glucose tolerance/insulin sensitivity in ob/ob mice. Taken together, S1pr2 blockade induced proliferation but suppressed differentiation of (pre)adipocytes both in vivo and in vitro, highlighting a novel therapeutic approach for obesity/type 2 diabetes.


Assuntos
Adipócitos/metabolismo , Crescimento Celular/efeitos dos fármacos , Dieta Hiperlipídica , Intolerância à Glucose/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/genética , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Adipogenia/efeitos dos fármacos , Animais , Peso Corporal/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Intolerância à Glucose/genética , Masculino , Camundongos , Camundongos Knockout , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato
15.
PLoS One ; 10(12): e0144053, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26630567

RESUMO

Glucose is a primary stimulator of insulin secretion in pancreatic ß-cells. High concentration of glucose has been thought to exert its action solely through its metabolism. In this regard, we have recently reported that glucose also activates a cell-surface glucose-sensing receptor and facilitates its own metabolism. In the present study, we investigated whether glucose activates the glucose-sensing receptor and elicits receptor-mediated rapid actions. In MIN6 cells and isolated mouse ß-cells, glucose induced triphasic changes in cytoplasmic Ca(2+) concentration ([Ca(2+)]c); glucose evoked an immediate elevation of [Ca(2+)]c, which was followed by a decrease in [Ca(2+)]c, and after a certain lag period it induced large oscillatory elevations of [Ca(2+)]c. Initial rapid peak and subsequent reduction of [Ca(2+)]c were independent of glucose metabolism and reproduced by a nonmetabolizable glucose analogue. These signals were also blocked by an inhibitor of T1R3, a subunit of the glucose-sensing receptor, and by deletion of the T1R3 gene. Besides Ca(2+), glucose also induced an immediate and sustained elevation of intracellular cAMP ([cAMP]c). The elevation of [cAMP]c was blocked by transduction of the dominant-negative Gs, and deletion of the T1R3 gene. These results indicate that glucose induces rapid changes in [Ca(2+)]c and [cAMP]c by activating the cell-surface glucose-sensing receptor. Hence, glucose generates rapid intracellular signals by activating the cell-surface receptor.


Assuntos
Cálcio/metabolismo , AMP Cíclico/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Linhagem Celular , Citoplasma/metabolismo , Insulina/metabolismo , Camundongos
16.
Biol Pharm Bull ; 38(5): 674-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25947913

RESUMO

Subunits of the sweet taste receptors T1R2 and T1R3 are expressed in pancreatic ß-cells. Compared with T1R3, mRNA expression of T1R2 is considerably lower. At the protein level, expression of T1R2 is undetectable in ß-cells. Accordingly, a major component of the sweet taste-sensing receptor in ß-cells may be a homodimer of T1R3 rather than a heterodimer of T1R2/T1R3. Inhibition of this receptor by gurmarin or deletion of the T1R3 gene attenuates glucose-induced insulin secretion from ß-cells. Hence the T1R3 homodimer functions as a glucose-sensing receptor (GSR) in pancreatic ß-cells. When GSR is activated by the T1R3 agonist sucralose, elevation of intracellular ATP concentration ([ATP]i) is observed. Sucralose increases [ATP]i even in the absence of ambient glucose, indicating that sucralose increases [ATP]i not simply by activating glucokinase, a rate-limiting enzyme in the glycolytic pathway. In addition, sucralose augments elevation of [ATP]i induced by methylsuccinate, suggesting that sucralose activates mitochondrial metabolism. Nonmetabolizable 3-O-methylglucose also increases [ATP]i and knockdown of T1R3 attenuates elevation of [ATP]i induced by high concentration of glucose. Collectively, these results indicate that the T1R3 homodimer functions as a GSR; this receptor is involved in glucose-induced insulin secretion by activating glucose metabolism probably in mitochondria.


Assuntos
Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sacarose/análogos & derivados , Paladar , 3-O-Metilglucose/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Camundongos , Mitocôndrias/metabolismo , Sacarose/farmacologia , Edulcorantes/farmacologia
17.
J Endocrinol ; 226(1): 57-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25994004

RESUMO

Glucose activates the glucose-sensing receptor T1R3 and facilitates its own metabolism in pancreatic ß-cells. An inhibitor of this receptor would be helpful in elucidating the physiological function of the glucose-sensing receptor. The present study was conducted to examine whether or not lactisole can be used as an inhibitor of the glucose-sensing receptor. In MIN6 cells, in a dose-dependent manner, lactisole inhibited insulin secretion induced by sweeteners, acesulfame-K, sucralose and glycyrrhizin. The IC50 was ∼4 mmol/l. Lactisole attenuated the elevation of cytoplasmic Ca2+ concentration ([Ca2+]c) evoked by sucralose and acesulfame-K but did not affect the elevation of intracellular cAMP concentration ([cAMP]c) induced by these sweeteners. Lactisole also inhibited the action of glucose in MIN6 cells. Thus, lactisole significantly reduced elevations of intracellular [NADH] and intracellular [ATP] induced by glucose, and also inhibited glucose-induced insulin secretion. To further examine the effect of lactisole on T1R3, we prepared HEK293 cells stably expressing mouse T1R3. In these cells, sucralose elevated both [Ca2+]c and [cAMP]c. Lactisole attenuated the sucralose-induced increase in [Ca2+]c but did not affect the elevation of [cAMP]c. Finally, lactisole inhibited insulin secretion induced by a high concentration of glucose in mouse islets. These results indicate that the mouse glucose-sensing receptor was inhibited by lactisole. Lactisole may be useful in assessing the role of the glucose-sensing receptor in mouse pancreatic ß-cells.


Assuntos
Derivados de Benzeno/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Edulcorantes/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Ácido Glicirrízico/farmacologia , Células HEK293 , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Sacarose/análogos & derivados , Sacarose/farmacologia , Tiazinas/farmacologia
18.
J Diabetes Investig ; 6(3): 256-63, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25969708

RESUMO

Subunits of the sweet taste receptor, namely T1R2 and T1R3, are expressed in mouse pancreatic islets. Quantitatively, the expression of messenger ribonucleic acid for T1R2 is much lower than that of T1R3, and immunoreactive T1R2 is in fact undetectable. Presumably, a homodimer of T1R3 could function as a signaling receptor. Activation of this receptor by adding an artificial sweetener, sucralose, leads to an increase in intracellular adenosine triphosphate ([ATP]c). This increase in [ATP]c is observed in the absence of ambient glucose. Sucralose also augments elevation of [ATP]c induced by methylsuccinate, a substrate for mitochondria. Consequently, activation of T1R3 promotes metabolism in mitochondria and increases [ATP]c. 3-O-Methylglucose, a non-metabolizable analog of glucose, also increases [ATP]c. Conversely, knockdown of T1R3 attenuates elevation of [ATP]c induced by glucose. Hence, glucose promotes its own metabolism by activating T1R3 and augmenting ATP production. Collectively, a homodimer of T1R3 functions as a cell surface glucose-sensing receptor and participates in the action of glucose on insulin secretion. The glucose-sensing receptor T1R3 might be the putative glucoreceptor proposed decades ago by Niki et al. The glucose-sensing receptor is involved in the action of glucose and modulates glucose metabolism in pancreatic ß-cells.

19.
Physiol Rep ; 3(2)2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25677550

RESUMO

The effect of focal mechanical stress on the localization of TRPV2 was investigated in HT1080 cells, where only mRNA for TRPV2 was detected among members of the TRPV channel family. Mechanical stress was applied by adding negative pressure using a glass pipette. When focal mechanical stress was applied, subplasma membrane Ca(2+) concentration ([Ca(2+)]s) was increased beneath the pipette, which propagated throughout the cell. The increase in [Ca(2+)]s was blocked by ruthenium red or by knocking down TRPV2. Elevation of [Ca(2+)]s was not observed by removal of extracellular Ca(2+), by an addition of a phosphatidylinositol 3-kinase inhibitor LY29034, and by transfection of dominant-negative Rac. In cells expressing GFP-TRPV2 and RFP-Akt, administration of focal mechanical stress induced accumulation of GFP-TRPV2 beneath the pipette. RFP-Akt was also accumulated to the same site. Gadolinium blocked the elevation of [Ca(2+)]s induced by focal mechanical stress and also attenuated accumulation of TRPV2. When GFP-TRPV1, GFP-TRPV3, GFP-TRPV4, GFP-TRPV5, or GFP-TRPV6 was transfected ectopically in HT1080 cells, only GFP-TRPV4 was accumulated beneath the pipette in response to the focal mechanical stress. These results indicate that TRPV2 translocates to the site receiving a focal mechanical stress and increases [Ca(2+)]s.

20.
J Hepatobiliary Pancreat Sci ; 21(12): 856-63, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25139445

RESUMO

BACKGROUND: The aim of the present study was to clarify the anatomy between the left triangular ligament (LTL) and the appendix fibrosa hepatis (AFH) in order not to sever the AFH when dissecting the LTL. METHODS: Totals of 43 and 27 cadaveric livers were examined macroscopically and histologically, respectively. RESULTS: The LTL attached itself to the diaphragmatic surface of the AFH through almost all lengths of the AFH. This might be the reason why AFH is so often dissected together with the LTL. There were two types of relation between the LTL and the AFH; in one type, the starting point of the LTL existed on the left liver and in the other type, it was on the AFH. Twenty-five of 27 AFH included remnants of the bile duct and 12 of 25 AFH had comparatively large bile ducts, which was unexceptionally accompanied by the well-developed peribiliary vascular plexus. AFH showed a variety of shapes, such as rectangular (6/43), long triangular (4/43), short triangular (7/43), triangular plus cordlike (11/43), cordlike (12/43) and bifurcated (3/43) types. CONCLUSIONS: As AFH sometimes includes relatively large bile ducts, it is recommended for surgeons to sever the AFH not just simply by electrocautery but by ligating its stump securely.


Assuntos
Ligamentos/anatomia & histologia , Fígado/anatomia & histologia , Abdome/cirurgia , Idoso de 80 Anos ou mais , Ductos Biliares/anatomia & histologia , Cadáver , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...