Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(7): e0182256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28753672

RESUMO

A compensatory increase in ß-cell mass occurs during pregnancy to counter the associated insulin resistance, and a failure in adaptation is thought to contribute to gestational diabetes. Insulin-expressing but glucose-transporter-2-low (Ins+Glut2LO) progenitor cells are present in mouse and human pancreas, being predominantly located in extra-islet ß-cell clusters, and contribute to the regeneration of the endocrine pancreas following induced ablation. We therefore sought to investigate the contribution of Ins+Glut2LO cells to ß-cell mass expansion during pregnancy. Female C57Bl/6 mice were time mated and pancreata were collected at gestational days (GD) 6, 9, 12, 15, and 18, and postpartum D7 (n = 4/time-point) and compared to control (non-pregnant) animals. Beta cell mass, location, proliferation (Ki67+), and proportion of Ins+Glut2LO cells were measured using immunohistochemistry and bright field or confocal microscopy. Beta cell mass tripled by GD18 and ß-cell proliferation peaked at GD12 in islets (≥6 ß-cells) and small ß-cell clusters (1-5 ß-cells). The proportion and fraction of Ins+Glut2LO cells undergoing proliferation increased significantly at GD9 in both islets and clusters, preceding the increase in ß-cell mass and proliferation, and their proliferation within clusters persisted until GD15. The overall number of clusters increased significantly at GD9. Quantitative PCR showed a significant increase in Pdx1 presence at GD9 vs. GD18 or control pancreas, and Pdx1 was visualized by immunohistochemistry within both Ins+Glut2LO and Ins+Glut2HI cells within clusters. These results indicate that Ins+Glut2LO cells are likely to contribute to ß-cell mass expansion during pregnancy.


Assuntos
Diabetes Gestacional/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Feminino , Idade Gestacional , Transportador de Glucose Tipo 2/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Gravidez
2.
Nephrol Dial Transplant ; 29 Suppl 4: iv113-6, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25165176

RESUMO

Fanconi-Bickel syndrome is a rare autosomal-recessive disorder caused by mutations in the SLC2A2 gene coding for the glucose transporter protein 2 (GLUT2). Major manifestations include hepatomegaly, glucose intolerance, post-prandial hypoglycaemia and renal disease that usually presents as proximal tubular acidosis associated with proximal tubule dysfunction (renal Fanconi syndrome). We report a patient harbouring a homozygous mutation of SLC2A2 who presented a dramatic exacerbation of metabolic acidosis in the context of a viral infection, owing to both ketosis and major urinary bicarbonate loss. The kidney biopsy revealed nuclear and cytoplasmic accumulation of glycogen in proximal tubule cells, a lack of expression of GLUT2, and major defects of key proteins of the proximal tubule such as megalin, cubilin and the B2 subunit of H(+)-ATPase. These profound alterations of the transport systems most likely contributed to proximal tubule alterations and profound bicarbonate loss.


Assuntos
Acidose/fisiopatologia , Síndrome de Fanconi/fisiopatologia , Transportador de Glucose Tipo 2/genética , Túbulos Renais Proximais/fisiopatologia , Mutação/genética , Acidose/genética , Doença Aguda , Síndrome de Fanconi/genética , Transportador de Glucose Tipo 2/deficiência , Homozigoto , Humanos , Lactente , Masculino
3.
PLoS One ; 9(2): e89977, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587162

RESUMO

Intestinal glucose absorption is mediated by SGLT1 whereas GLUT2 is considered to provide basolateral exit. Recently, it was proposed that GLUT2 can be recruited into the apical membrane after a high luminal glucose bolus allowing bulk absorption of glucose by facilitated diffusion. Moreover, SGLT1 and GLUT2 are suggested to play an important role in intestinal glucose sensing and incretin secretion. In mice that lack either SGLT1 or GLUT2 we re-assessed the role of these transporters in intestinal glucose uptake after radiotracer glucose gavage and performed Western blot analysis for transporter abundance in apical membrane fractions in a comparative approach. Moreover, we examined the contribution of these transporters to glucose-induced changes in plasma GIP, GLP-1 and insulin levels. In mice lacking SGLT1, tissue retention of tracer glucose was drastically reduced throughout the entire small intestine whereas GLUT2-deficient animals exhibited higher tracer contents in tissue samples than wild type animals. Deletion of SGLT1 resulted also in reduced blood glucose elevations and abolished GIP and GLP-1 secretion in response to glucose. In mice lacking GLUT2, glucose-induced insulin but not incretin secretion was impaired. Western blot analysis revealed unchanged protein levels of SGLT1 after glucose gavage. GLUT2 detected in apical membrane fractions mainly resulted from contamination with basolateral membranes but did not change in density after glucose administration. SGLT1 is unequivocally the prime intestinal glucose transporter even at high luminal glucose concentrations. Moreover, SGLT1 mediates glucose-induced incretin secretion. Our studies do not provide evidence for GLUT2 playing any role in either apical glucose influx or incretin secretion.


Assuntos
Transportador de Glucose Tipo 2/metabolismo , Glucose/metabolismo , Mucosa Intestinal/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Glicemia/metabolismo , Glucose/farmacologia , Transportador de Glucose Tipo 2/deficiência , Incretinas/sangue , Incretinas/metabolismo , Insulina/sangue , Insulina/metabolismo , Secreção de Insulina , Absorção Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Camundongos , Transportador 1 de Glucose-Sódio/deficiência
4.
J Clin Invest ; 124(1): 413-24, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24334455

RESUMO

How glucose sensing by the nervous system impacts the regulation of ß cell mass and function during postnatal development and throughout adulthood is incompletely understood. Here, we studied mice with inactivation of glucose transporter 2 (Glut2) in the nervous system (NG2KO mice). These mice displayed normal energy homeostasis but developed late-onset glucose intolerance due to reduced insulin secretion, which was precipitated by high-fat diet feeding. The ß cell mass of adult NG2KO mice was reduced compared with that of WT mice due to lower ß cell proliferation rates in NG2KO mice during the early postnatal period. The difference in proliferation between NG2KO and control islets was abolished by ganglionic blockade or by weaning the mice on a carbohydrate-free diet. In adult NG2KO mice, first-phase insulin secretion was lost, and these glucose-intolerant mice developed impaired glucagon secretion when fed a high-fat diet. Electrophysiological recordings showed reduced parasympathetic nerve activity in the basal state and no stimulation by glucose. Furthermore, sympathetic activity was also insensitive to glucose. Collectively, our data show that GLUT2-dependent control of parasympathetic activity defines a nervous system/endocrine pancreas axis that is critical for ß cell mass establishment in the postnatal period and for long-term maintenance of ß cell function.


Assuntos
Proliferação de Células , Transportador de Glucose Tipo 2/genética , Glucose/metabolismo , Homeostase , Células Secretoras de Insulina/metabolismo , Potenciais de Ação , Animais , Fibras Autônomas Pré-Ganglionares/fisiologia , Metabolismo Energético , Feminino , Gânglios Parassimpáticos/metabolismo , Gânglios Parassimpáticos/fisiopatologia , Intolerância à Glucose/metabolismo , Transportador de Glucose Tipo 2/deficiência , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/inervação , Pâncreas/patologia
5.
J Child Neurol ; 27(6): 796-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22156785

RESUMO

Mitochondrial disorders are varied in their clinical presentation and pathogenesis. Diagnosis is usually made clinically and genetic defects are often not identified. We present a 6-year-old female patient with a diagnosis of a mitochondrial disorder secondary to complex I deficiency with seizures and developmental delay from infancy. Glucose transporter deficiency was suspected after a lumbar puncture showed hypoglycorrhachia. Her disorder was confirmed genetically as a mutation in her solute carrier family 2, facilitated glucose transporter member 1 (SLCA2) gene. Delayed diagnosis led to delayed treatment, and neurologic sequelae may have been prevented by earlier recognition of this disorder.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/complicações , Erros Inatos do Metabolismo dos Carboidratos/genética , Deficiências do Desenvolvimento/genética , Transportador de Glucose Tipo 2/deficiência , Doenças Mitocondriais/etiologia , Doenças Mitocondriais/genética , Erros Inatos do Metabolismo dos Carboidratos/líquido cefalorraquidiano , Criança , Deficiências do Desenvolvimento/líquido cefalorraquidiano , Deficiências do Desenvolvimento/complicações , Feminino , Glucose/líquido cefalorraquidiano , Humanos , Doenças Mitocondriais/líquido cefalorraquidiano , Punção Espinal
6.
IUBMB Life ; 62(5): 315-33, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20209635

RESUMO

The protein family of facilitative glucose transporters comprises 14 isoforms that share common structural features such as 12 transmembrane domains, N- and C-termini facing the cytoplasm of the cell, and a N-glycosylation side either within the first or fifth extracellular loop. Based on their sequence homology, three classes can be distinguished: class I includes GLUT1-4 and GLUT14, class II the "odd transporters" GLUT5, 7, 9, 11, and class III the "even transporters" GLUT6, 8, 10, 12 and the proton driven myoinositol transporter HMIT (or GLUT13). With the cloning and characterization of the more recent class II and III isoforms, it became apparent that despite their structural similarities, the different isoforms not only show a distinct tissue-specific expression pattern but also show distinct characteristics such as alternative splicing, specific (sub)cellular localization, and affinities for a spectrum of substrates. This review summarizes the current understanding of the physiological role for the various transport facilitators based on human genetically inherited disorders or single-nucleotide polymorphisms and knockout mice models. The emphasis of the review will be on the potential functional role of the more recent isoforms.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/fisiologia , Sequência de Aminoácidos , Animais , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportador de Glucose Tipo 1/fisiologia , Transportador de Glucose Tipo 2/deficiência , Transportador de Glucose Tipo 2/metabolismo , Transportador de Glucose Tipo 3/fisiologia , Transportador de Glucose Tipo 4/metabolismo , Transportador de Glucose Tipo 4/fisiologia , Transportador de Glucose Tipo 5/fisiologia , Humanos , Camundongos , Filogenia , Especificidade por Substrato
7.
Cell Metab ; 8(3): 201-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18762021

RESUMO

Unlike the adjustable gastric banding procedure (AGB), Roux-en-Y gastric bypass surgery (RYGBP) in humans has an intriguing effect: a rapid and substantial control of type 2 diabetes mellitus (T2DM). We performed gastric lap-band (GLB) and entero-gastro anastomosis (EGA) procedures in C57Bl6 mice that were fed a high-fat diet. The EGA procedure specifically reduced food intake and increased insulin sensitivity as measured by endogenous glucose production. Intestinal gluconeogenesis increased after the EGA procedure, but not after gastric banding. All EGA effects were abolished in GLUT-2 knockout mice and in mice with portal vein denervation. We thus provide mechanistic evidence that the beneficial effects of the EGA procedure on food intake and glucose homeostasis involve intestinal gluconeogenesis and its detection via a GLUT-2 and hepatoportal sensor pathway.


Assuntos
Derivação Gástrica , Gastroplastia , Gluconeogênese , Glucose/metabolismo , Intestino Delgado/metabolismo , Obesidade Mórbida/metabolismo , Animais , Gorduras na Dieta/administração & dosagem , Ingestão de Alimentos , Transportador de Glucose Tipo 1/deficiência , Transportador de Glucose Tipo 2/deficiência , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Obesidade Mórbida/cirurgia , Veia Porta/metabolismo , Reprodutibilidade dos Testes , Fatores de Tempo
8.
Mol Cell Endocrinol ; 276(1-2): 18-23, 2007 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-17681422

RESUMO

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretins secreted in response to oral glucose ingestion by intestinal L and K cells, respectively. The molecular mechanisms responsible for intestinal cell glucose sensing are unknown but could be related to those described for beta-cells, brain and hepatoportal sensors. We determined the role of GLUT2, GLP-1 or GIP receptors in glucose-induced incretins secretion, in the corresponding knockout mice. GLP-1 secretion was reduced in all mutant mice, while GIP secretion did not require GLUT2. Intestinal GLP-1 content was reduced only in GIP and GLUT2 receptors knockout mice suggesting that this impairment could contribute to the phenotype. Intestinal GIP content was similar in all mice studied. Furthermore, the impaired incretins secretion was associated with a reduced glucose-stimulated insulin secretion and an impaired glucose tolerance in all mice. In conclusion, both incretins secretion depends on mechanisms involving their own receptors and GLP-1 further requires GLUT2.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Glucose/farmacologia , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Glucagon/metabolismo , Animais , Receptor do Peptídeo Semelhante ao Glucagon 1 , Intolerância à Glucose , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 2/deficiência , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Modelos Biológicos , Sistema Porta/efeitos dos fármacos , Sistema Porta/metabolismo , Receptores dos Hormônios Gastrointestinais/deficiência , Receptores de Glucagon/deficiência
9.
Exp Clin Endocrinol Diabetes ; 114(5): 262-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16804801

RESUMO

The New Zealand obese mouse (NZO/Hl) is characterised by hereditary obesity and type-2 diabetes, including insulin resistance, hyperinsulinaemia, and glucose intolerance. In other diabetic models, it has been revealed that the proper functioning of the glucose transporter isoform 2 (GLUT2) is essential for adequate secretion of insulin. The aim of this study was to compare the distribution of islet cells and GLUT2, as well as the expression of GLUT2-mRNA, in the pancreas of NZO mice and metabolically unimpaired NMRI (Naval Medical Research Institute) mice. Pancreas tissue was obtained from different stages of development. For molecular determination of the expression level of GLUT2-mRNA, total-RNA was extracted from the pancreas and analysed by quantitative real-time RT-PCR. All investigated NZO mice displayed increased weight, elevated hyperinsulinaemia, and slightly enhanced blood glucose levels compared with the NMRI control mice. By means of immunofluorescence microscopy drastically reduced insulin levels were detected, which might be compensated by the observed islet cell hyperplasia and hypertrophy. Furthermore, the normally peripheral localisation of the alpha-cells within islets was disturbed. By contrast, there were no changes in somatostatin cell distribution. However, considerable differences appeared with regard to GLUT2: whereas the beta-cells of NMRI mice showed dense immunostaining of the GLUT2 transporter on the cell surface, in all age groups of NZO mice, GLUT2 on the plasma membranes was reduced and dispersed in the cytoplasm. These findings agree with the molecular biological results, which displayed decreased mRNA-expression of GLUT2. In summary, the observed alteration of islet morphology and of GLUT2 expression in diabetic mice complements our previous results from a superfusion protocol and further clarifies the mechanisms of diabetogenesis in NZO mice.


Assuntos
Diabetes Mellitus/genética , Transportador de Glucose Tipo 2/deficiência , Transportador de Glucose Tipo 2/genética , Ilhotas Pancreáticas/metabolismo , Perda de Heterozigosidade , Animais , Arginina/farmacologia , Glicemia/metabolismo , Constituição Corporal , Instabilidade Cromossômica , Diabetes Mellitus/patologia , Feminino , Expressão Gênica , Glucose/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Insulina/sangue , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Obesos , Pâncreas/metabolismo , RNA Mensageiro/metabolismo
10.
Diabetes ; 55(4): 988-95, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16567520

RESUMO

A role for glucose in the control of feeding has been proposed, but its precise physiological importance is unknown. Here, we evaluated feeding behavior in glut2-null mice, which express a transgenic glucose transporter in their beta-cells to rescue insulin secretion (ripglut1;glut2-/- mice). We showed that in the absence of GLUT2, daily food intake was increased and feeding initiation and termination following a fasting period were abnormal. This was accompanied by suppressed regulation of hypothalamic orexigenic and anorexigenic neuropeptides expression during the fast-to-refed transition. In these conditions, however, there was normal regulation of the circulating levels of insulin, leptin, or glucose but a loss of regulation of plasma ghrelin concentrations. To evaluate whether the abnormal feeding behavior was due to suppressed glucose sensing, we evaluated feeding in response to intraperitoneal or intracerebroventricular glucose or 2-deoxy-D-glucose injections. We showed that in GLUT2-null mice, feeding was no longer inhibited by glucose or activated by 2-deoxy-D-glucose injections and the regulation of hypothalamic neuropeptide expression by intracerebroventricular glucose administration was lost. Together, these data demonstrate that absence of GLUT2 suppressed the function of central glucose sensors, which control feeding probably by regulating the hypothalamic melanocortin pathway. Furthermore, inactivation of these glucose sensors causes overeating.


Assuntos
Comportamento Alimentar/fisiologia , Transportador de Glucose Tipo 2/fisiologia , Animais , Sequência de Bases , Primers do DNA , Grelina , Transportador de Glucose Tipo 2/deficiência , Transportador de Glucose Tipo 2/genética , Hipotálamo/fisiologia , Insulina/sangue , Leptina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/genética , Hormônios Peptídicos/sangue , Pró-Opiomelanocortina/genética , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , alfa-MSH/fisiologia
11.
Mol Genet Metab ; 86(4): 473-7, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16288895

RESUMO

The Fanconi-Bickel syndrome is caused by homozygosity or compound heterozygosity for mutations of the facilitated glucose transporter 2 gene (GLUT2). Glycogen accumulates in renal tubular cells and they fail to reabsorb multiple filtered solutes because of impairment in GLUT2-mediated efflux of glucose. We describe a 10-year-old male child with GLUT2 deficiency who produced massive amounts of 3-deoxyfructose (3-DF) in the kidneys. Since 3-DF is a detoxification product of a potent glycating agent, 3-deoxyglucosone, a precursor of advanced glycation end-products, this suggests a massive accumulation of glucose within tubular cells probably as a consequence of GLUT2 deficiency. The level of 3-DF in the urine of this atypical patient, who also manifested renal glomerular hyperfiltration, microalbuminuria, and glomerular mesangial expansion, was higher than in any patient examined with diabetes mellitus. Elevated levels of glucose and/or its metabolites in renal tubular cells may be necessary but not sufficient for the development of both the renal tubulopathy and diabetic-like glomerular disease in GLUT2 deficiency.


Assuntos
Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Síndrome de Fanconi/complicações , Síndrome de Fanconi/genética , Transportador de Glucose Tipo 2/deficiência , Transportador de Glucose Tipo 2/genética , Adulto , Estudos de Casos e Controles , Criança , Análise Mutacional de DNA , Nefropatias Diabéticas/metabolismo , Síndrome de Fanconi/metabolismo , Humanos , Cetoses/urina , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...