Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
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.
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
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.
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
5.
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
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.
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
8.
J Biol Chem ; 289(1): 133-42, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24240093

RESUMO

Although insulin acutely stimulates glucose uptake by promotion of GLUT4 translocation from intracellular compartments to the plasma membrane in adipocytes and muscles, long term insulin stimulation causes GLUT4 depletion that is particularly prominent in the insulin-responsive GLUT4 storage compartment. This effect is caused mainly by accelerated lysosomal degradation of GLUT4, although the mechanism is not fully defined. Here we show that insulin acutely induced dissociation of retromer components from the low density microsomal membranes of 3T3-L1 adipocytes that was accompanied by disruption of the interaction of Vps35 with sortilin. This insulin effect was dependent on the activity of protein kinase CK2 but not phosphatidylinositol 3-kinase or extracellular signal-regulated kinase 1/2. Knockdown of Vps26 decreased GLUT4 to a level comparable with that with insulin stimulation for 4 h. Vps35 with a mutation in the CK2 phosphorylation motif (Vps35-S7A) was resistant to insulin-induced dissociation from the low density microsomal membrane, and its overexpression attenuated GLUT4 down-regulation with insulin. Furthermore, insulin-generated hydrogen peroxide was an upstream mediator of the insulin action on retromer and GLUT4. These results suggested that insulin-generated oxidative stress switches the GLUT4 sorting direction to lysosomes through inhibition of the retromer function in a CK2-dependent manner.


Assuntos
Adipócitos/metabolismo , Caseína Quinase II/metabolismo , Regulação para Baixo/fisiologia , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Estresse Oxidativo/fisiologia , Células 3T3 , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Adipócitos/citologia , Animais , Caseína Quinase II/genética , Regulação para Baixo/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Transportador de Glucose Tipo 4/genética , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Membranas Intracelulares/metabolismo , Lipossomos/metabolismo , Camundongos , Microssomos/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Proteólise/efeitos dos fármacos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
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
10.
Mol Cell Biochem ; 391(1-2): 217-24, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24604674

RESUMO

Thyroid hormone stimulates erythropoietic differentiation. However, severe anemia is sometimes seen in patients with hyperthyroidism, and the mechanisms have not been fully elucidated. Bone marrow is comprised about 2-8% oxygen, and the characteristics of hematopoietic stem cells have been shown to be influenced under hypoxia. Hypoxia-inducible factor-1 is a critical mediator of cellular responses to hypoxia and an important mediator in signal transduction of thyroid hormone [triiodothyronine (T3)]. The aim of this study was to investigate the effect of T3 on erythropoiesis under hypoxia mimicking physiological conditions in the bone marrow. We maintained human erythroleukemia K562 cells under hypoxic atmosphere (2% O2) and examined their cellular characteristics. Compared to that under normal atmospheric conditions, cells under hypoxia showed a reduction in the proliferation rate and increase in the hemoglobin content or benzidine-positive rate, indicating promotion of erythroid differentiation. T3 had no effect on hypoxia-induced erythroid differentiation, but significantly inhibited activin A/erythroid differentiation factor-induced erythroid differentiation. Moreover, GATA2 mRNA expression was suppressed in association with erythroid differentiation, while T3 significantly diminished that suppression. These results suggest that T3 has a direct suppressive effect on erythroid differentiation under hypoxic conditions.


Assuntos
Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Leucemia Eritroblástica Aguda/patologia , Tri-Iodotironina/farmacologia , Hipóxia Celular/efeitos dos fármacos , Feminino , Humanos , Células K562 , Pessoa de Meia-Idade
11.
Liver Int ; 34(7): 1057-67, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24119135

RESUMO

BACKGROUND & AIMS: Conophylline (CnP) is a vinca alkaloid purified from a tropical plant and inhibits activation of pancreatic stellate cells. We investigated the effect of CnP on hepatic stellate cells (HSC) in vitro. We also examined whether CnP attenuates hepatic fibrosis in vivo. METHOD: We examined the effect of CnP on the expression of α-smooth muscle actin (α-SMA) and collagen-1, DNA synthesis and apoptosis in rat HSC and Lx-2 cells. We also examined the effect of CnP on hepatic fibrosis induced by thioacetamide (TAA). RESULTS: In rat HSC and Lx-2 cells, CnP reduced the expression of α-SMA and collagen-1. CnP inhibited DNA synthesis induced by serum. CnP also promoted activation of caspase-3 and induced apoptosis as assessed by DNA ladder formation and TUNEL assay. In contrast, CnP did not induce apoptosis in AML12 cells. We then examined the effect of CnP on TAA-induced cirrhosis. In TAA-treated rats, the surface of the liver was irregular and multiple nodules were observed. Histologically, formation of pseudolobules surrounded by massive fibrous tissues was observed. When CnP was administered together with TAA, the surface of the liver was smooth and liver fibrosis was markedly inhibited. Collagen content was significantly reduced in CnP-treated liver. CONCLUSION: Conophylline suppresses HSC and induces apoptosis in vitro. CnP also attenuates formation of the liver fibrosis induced by TAA in vivo.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Tioacetamida/efeitos adversos , Alcaloides de Vinca/farmacologia , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Linhagem Celular , Colágeno Tipo I/metabolismo , Replicação do DNA/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Ratos , Alcaloides de Vinca/uso terapêutico
12.
Endocr J ; 61(2): 119-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24200979

RESUMO

A homodimer of taste type 1 receptor 3 (T1R3) functions as a sweet taste-sensing receptor in pancreatic ß-cells. This receptor is activated by various sweet molecules including sugars such as glucose. To determine the role of this receptor in glucose-induced insulin secretion, we addressed whether or not this receptor modulates glucose metabolism in MIN6 cells. We measured changes in intracellular ATP ([ATP]i) in MIN6 cells expressing luciferase. Sucralose, an agonist of T1R3, induced immediate and sustained elevation of [ATP]i in the presence of 5.5 mM glucose. The effect of sucralose was dose-dependent and, at 5 mM, was greater than that induced by 25 mM glucose. In contrast, carbachol, GLP-1 or high concentration of potassium did not reproduce the sucralose action. Sucralose facilitated the increase in [ATP]i induced by a mitochondrial fuel methylsuccinate, and potentiated glucose-induced elevation of [ATP]i. Administration of a non-metabolizable glucose analogue, 3-O-methylglucose, which acts as an agonist of T1R3, induced a small and transient increase in [ATP]i. 3-O-Methylglucose augmented elevation of [ATP]i induced by methylsuccinate, and also enhanced glucose-induced increase in [ATP]i. Knock down of T1R3 by using shRNA attenuated [ATP]i-response to high concentration of glucose and also reduced the glucose-induced insulin secretion. These results indicate that activation of the homodimer of T1R3 facilitates the metabolic pathway in mitochondria and augments ATP production. The results obtained by using 3-O-methylglucose suggest that glucose, by acting on the homodimer of T1R3, promotes its own metabolism.


Assuntos
Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , 3-O-Metilglucose/farmacologia , Animais , Linhagem Celular , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Succinatos/farmacologia , Sacarose/análogos & derivados , Sacarose/farmacologia
13.
Endocr J ; 61(8): 797-805, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24898279

RESUMO

We reported recently that the taste type 1 receptor 3 (T1R3), a subunit of the sweet taste receptor, functions as a cell-surface glucose-sensing receptor in pancreatic ß-cells. In the present study, we investigated the expression of T1R3 in pancreatic islets. mRNA for T1R2 and T1R3 was detected in mouse pancreatic islets. Quantitatively, the mRNA expression level of T1R2 was less than 1% of that of T1R3. Immunohistochemically, T1R3 was abundantly expressed in mouse islets whereas T1R2 was barely detected. Most immunoreactive T1R3 was colocalized with insulin and almost all ß-cells were positive for T1R3. In addition, T1R3 was expressed in some portion of α-cells. Immunoreactivity of T1R3 in ß-cells was markedly reduced in fed mice compared to those in fasting mice. In contrast, mRNA for T1R3 was not different in islets of fasting and fed mice. Glucose-induced insulin-secretion was higher in islets obtained from fasting mice compared to those from fed mice. The expression of T1R3 was markedly reduced in islets of ob/ob mice compared to those of control mice. Similarly, the expression of T1R3 was reduced in islet of db/db mice. In addition, the expression of T1R3 was markedly reduced in ß-cells of fatty diabetic rats and GK rats, models of obese and non-obese type 2 diabetes, respectively. These results indicate that T1R3 is expressed mainly in ß-cells and the expression levels are different depending upon the nutritional and metabolic conditions.


Assuntos
Metabolismo Energético/fisiologia , Ilhotas Pancreáticas/metabolismo , Estado Nutricional/fisiologia , Receptores Acoplados a Proteínas G/genética , Animais , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Transgênicos , Ratos Wistar , Receptores Acoplados a Proteínas G/metabolismo
14.
Handb Exp Pharmacol ; 222: 247-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24756709

RESUMO

Transient receptor potential vanilloid type 2, TRPV2, is a calcium-permeable cation channel belonging to the TRPV channel family. This channel is activated by heat (>52 °C), various ligands, and mechanical stresses. In most of the cells, a large portion of TRPV2 is located in the endoplasmic reticulum under unstimulated conditions. Upon stimulation of the cells with phosphatidylinositol 3-kinase-activating ligands, TRPV2 is translocated to the plasma membrane and functions as a cation channel. Mechanical stress may also induce translocation of TRPV2 to the plasma membrane. The expression of TRPV2 is high in some types of cells including neurons, neuroendocrine cells, immune cells involved in innate immunity, and certain types of cancer cells. TRPV2 may modulate various cellular functions in these cells.


Assuntos
Canais de Cálcio/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Canais de Cálcio/química , Canais de Cálcio/deficiência , Canais de Cálcio/genética , Permeabilidade da Membrana Celular , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Ativação do Canal Iônico , Potenciais da Membrana , Camundongos Knockout , Fenótipo , Transdução de Sinais , Canais de Cátion TRPV/química , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética
15.
Endocr J ; 60(5): 687-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23411584

RESUMO

The mechanism that initiates regeneration of pancreatic ß-cells is not clear at present. The vagal nerve is implicated in the regulation of gastrointestinal functions, glucose metabolism and proliferation of pancreatic ß-cells under physiological conditions. To elucidate the triggering mechanism of the regeneration of pancreatic ß-cells, we examined the involvement of the vagal nerve. To this end, we employed a rat pancreatic duct ligation (DL) model, in which profound ß-cell neogenesis and ß-cell proliferation were observed within a week. We administered atropine to block the vagal nerve. Administration of atropine inhibited proliferation of ß-cells in both islets and islet-like cell clusters (ICC), without affecting ductal cell proliferation in the ligated pancreas. The numbers of PDX-1 and MafB-positive cells in or attaching to the ducts were significantly reduced by atropine. MafB/glucagon and MafB/insulin double-positive cells were also decreased by atropine. Finally, atropine reduced the number of MafA-positive ductal cells, all of which were positive for insulin, by 50% on day 5. These results strongly suggest that the vagal nerve is involved in ß-cell proliferation, induction of endocrine progenitors and neogenesis of α- and ß-cells.


Assuntos
Células Secretoras de Insulina/fisiologia , Pâncreas/inervação , Sistema Nervoso Parassimpático/fisiologia , Regeneração , Células-Tronco/citologia , Animais , Atropina/farmacologia , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Glucagon/metabolismo , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ligadura , Fator de Transcrição MafB/metabolismo , Masculino , Proteínas Oncogênicas/metabolismo , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Ductos Pancreáticos/citologia , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/cirurgia , Sistema Nervoso Parassimpático/citologia , Sistema Nervoso Parassimpático/efeitos dos fármacos , Parassimpatolíticos/farmacologia , Ratos , Ratos Wistar , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Transativadores/metabolismo , Nervo Vago/citologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
16.
Endocr J ; 60(10): 1191-206, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23933592

RESUMO

The sweet taste receptor is expressed in the taste bud and is activated by numerous sweet molecules with diverse chemical structures. It is, however, not known whether these sweet agonists induce a similar cellular response in target cells. Using MIN6 cells, a pancreatic ß-cell line expressing endogenous sweet taste receptor, we addressed this question by monitoring changes in cytoplasmic Ca2+ ([Ca2+]i) and cAMP ([cAMP]i) induced by four sweet taste receptor agonists. Glycyrrhizin evoked sustained elevation of [Ca2+]i but [cAMP]i was not affected. Conversely, an artificial sweetener saccharin induced sustained elevation of [cAMP]i but did not increase [Ca2+]i. In contrast, sucralose and acesulfame K induced rapid and sustained increases in both [Ca2+]i and [cAMP]i. Although the latter two sweeteners increased [Ca2+]i and [cAMP]i, their actions were not identical: [Ca2+]i response to sucralose but not acesulfame K was inhibited by gurmarin, an antagonist of the sweet taste receptor which blocks the gustducin-dependent pathway. In addition, [Ca2+]i response to acesulfame K but not to sucralose was resistant to a Gq inhibitor. These results indicate that four types of sweeteners activate the sweet taste receptor differently and generate distinct patterns of intracellular signals. The sweet taste receptor has amazing multimodal functions producing multiple patterns of intracellular signals.


Assuntos
Transdução de Sinais/efeitos dos fármacos , Edulcorantes/farmacologia , Papilas Gustativas/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Citoplasma/metabolismo , Ácido Glicirrízico/farmacologia , Células Secretoras de Insulina/metabolismo , Camundongos , Proteínas de Plantas/farmacologia , Sacarina/farmacologia , Sacarose/análogos & derivados , Paladar/efeitos dos fármacos , Papilas Gustativas/efeitos dos fármacos , Tiazinas/farmacologia
17.
Gastroenterology ; 141(3): 1102-1113.e1-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21699790

RESUMO

BACKGROUND & AIMS: Pancreatic acinar cells are used to study regulated exocytosis. We investigated the role of interferon regulatory factor-2 (IRF2) in exocytosis in pancreatic acinar cells. METHODS: Pancreas tissues from Irf2⁺/⁺, Irf2⁺/⁻), and Irf2⁻/⁻ mice were examined by microscopy, immunohistochemical, and immunoblot analyses; amylase secretion was quantified. We also compared salivary glands and pancreatic islets of Irf2⁻/⁻ mice with those of Irf2⁺/⁻ mice. To examine the effects of increased signaling by type I interferons, we studied pancreatic acini from Irf2⁻/⁻Ifnar1⁻/⁻ mice. The effect of IRF2 on amylase secretion was studied using an acinar cell line and a retroviral system. We studied expression of IRF2 in wild-type mice with cerulein-induced pancreatitis and changes in pancreatic tissue of Irf2⁻/⁻ mice, compared with those of Irf2⁺/⁻ mice. RESULTS: Irf2⁻/⁻ pancreas was white and opaque; numerous and wide-spread zymogen granules were observed throughout the cytoplasm, along with lack of fusion between zymogen granules and the apical membrane, lack of secretagogue-stimulated amylase secretion, and low serum levels of amylase and elastase-1, indicating altered regulation of exocytosis. The expression pattern of soluble N-ethylmaleimide-sensitive factor attachment protein receptors changed significantly, specifically in pancreatic acini, and was not rescued by disruption of type I interferon signaling. Down-regulation of IRF2 decreased amylase secretion in an acinar cell line. In mice with pancreatitis, levels of IRF2 were reduced. Irf2⁻/⁻ acini were partially resistant to induction of pancreatitis. CONCLUSIONS: IRF2 regulates exocytosis in pancreatic acinar cells; defects in this process might be involved in the early phases of acute pancreatitis.


Assuntos
Exocitose/fisiologia , Fator Regulador 2 de Interferon/fisiologia , Pâncreas Exócrino/patologia , Pâncreas Exócrino/fisiopatologia , Proteínas SNARE/fisiologia , Animais , Autofagia/fisiologia , Linhagem Celular , Células Cultivadas , Ceruletídeo/efeitos adversos , Modelos Animais de Doenças , Fator Regulador 2 de Interferon/genética , Interferons/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pancreatite/induzido quimicamente , Pancreatite/patologia , Pancreatite/fisiopatologia , Transdução de Sinais/fisiologia
18.
Biochim Biophys Acta ; 1798(5): 966-74, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20067760

RESUMO

The pore-forming component of voltage-gated calcium channels, alpha(1) subunit, contains four structurally conserved domains (I-IV), each of which contains six transmembrane segments (S1-S6). We have shown previously that a Gly residue in the S2-S3 linker of domain III is completely conserved from yeasts to humans and important for channel activity. The Gly residues in the S2-S3 linkers of domains I and II, which correspond positionally to the Gly in the S2-S3 linker of domain III, are also highly conserved. Here, we investigated the role of the Gly residues in the S2-S3 linkers of domains I and II of Ca(v)1.2. Each of the Gly residues was replaced with Glu or Gln to produce mutant Ca(v)1.2s; G182E, G182Q, G579E, G579Q, and the resulting mutants were transfected into BHK6 cells. Whole-cell patch-clamp recordings showed that current-voltage relationships of the four mutants were the same as those of wild-type Ca(v)1.2. However, G182E and G182Q showed significantly smaller current densities because of mislocalization of the mutant proteins, suggesting that Gly(182) in domain I is involved in the membrane trafficking or surface expression of alpha(1) subunit. On the other hand, G579E showed a slower voltage-dependent current inactivation (VDI) compared to Ca(v)1.2, although G579Q showed a normal VDI, implying that Gly(579) in domain II is involved in the regulation of VDI and that the incorporation of a negative charge alters the VDI kinetics. Our findings indicate that the two conserved Gly residues are important for alpha(1) subunit to become functional.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Glicina/genética , Subunidades Proteicas/metabolismo , Sequência de Aminoácidos , Animais , Canais de Cálcio Tipo L/genética , Linhagem Celular , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Glicina/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Prolina/metabolismo , Subunidades Proteicas/genética , Ratos , Alinhamento de Sequência
19.
J Cell Biochem ; 112(1): 318-29, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21069736

RESUMO

Extracellular matrix (ECM) modulates differentiation of pancreatic ß-cells during development. However, the mechanism by which ECM proteins modulate differentiation is not totally clear. We investigated the effect of ECM proteins on differentiation ß-cells in vitro. We investigated the effect of basement membrane ECM on differentiation of AR42J cells and rat ductal cells. First, we examined the effect of reconstituted basement membrane, Matrigel on differentiation of AR42J cells induced by activin and betacellulin. Matrigel augmented insulin production and increased the expression of GLUT2, SUR1, and glucokinase. Among various transcription factors investigated, Matrigel markedly upregulated the expression of Pax6. When Pax6 was overexpressed in cells treated with activin and betacellulin, the expression of insulin was upregulated. Conversely, knockdown of Pax6 significantly reduced the insulin expression in cells cultured on Matrigel. The effects of Matrigel on insulin-production and induction of Pax6 were reproduced partially by laminin-1, a major component of Matrigel, and inhibited by anti-integrin-ß1 antibody. Matrigel also enhanced the activation of p38 mitogen-activated kinase induced by activin and betacellulin, which was inhibited by anti-ß1 antibody. Finally, the effect of Matrigel on differentiation was reproduced in rat cultured ductal cells, and Matrigel also increased the expression of Pax6. These results indicate that basement membrane ECM augments differentiation of pancreatic progenitor cells to insulin-secreting cells by upregulating the expression of Pax6. .


Assuntos
Diferenciação Celular , Matriz Extracelular/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Insulina/biossíntese , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular , Fator de Transcrição PAX6 , Pâncreas/citologia , Pâncreas/metabolismo , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Biochem Biophys Res Commun ; 416(1-2): 58-63, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22079287

RESUMO

GPR119 is one of the G-protein-coupled receptors expressed in pancreatic ß-cells and intestinal endocrine cells. Since agonists to GPR119 stimulate glucose-dependent insulin secretion, GPR119 agonists are anticipated to promote anti-diabetic effects and control of glucose homeostasis. Here, we reported that an omega-3 unsaturated fatty acid metabolite, 5-hydroxy-eicosapentaenoic acid (5-HEPE), was a potent agonist for GPR119 and enhanced glucose-dependent insulin secretion. 5-HEPE stimulated cAMP accumulation in mouse MIN6 insulinoma cells and human HuTu80 intestinal adenocarcinoma cells. These effects were blunted by GPR119-specific siRNA. Recombinant GPR119 also responded to 5-HEPE as well as authentic agonists. Several previous reports have indicated the beneficial biological effects of omega-3 unsaturated fatty acids, and epidemiological studies have suggested that these fatty acids plays a protective role against diabetes. However, the molecular pharmacology and receptor identifications of omega-3 unsaturated fatty acids and their metabolites have not yet been well investigated. It is hoped that our findings will encourage novel investigations into the molecular relationships between omega-3 fatty acids and diabetes.


Assuntos
Ácido Eicosapentaenoico/análogos & derivados , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Diabetes Mellitus/metabolismo , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacologia , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/metabolismo , Secreção de Insulina , Camundongos , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA