Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Osteoporos Int ; 26(1): 209-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25127672

ABSTRACT

UNLABELLED: A role for gut hormone in bone physiology has been suspected. We evidenced alterations of microstructural morphology (trabecular and cortical) and bone strength (both at the whole-bone--and tissue-level) in double incretin receptor knock-out (DIRKO) mice as compared to wild-type littermates. These results support a role for gut hormones in bone physiology. INTRODUCTION: The two incretins, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), have been shown to control bone remodeling and strength. However, lessons from single incretin receptor knock-out mice highlighted a compensatory mechanism induced by elevated sensitivity to the other gut hormone. As such, it is unclear whether the bone alterations observed in GIP or GLP-1 receptor deficient animals resulted from the lack of a functional gut hormone receptor, or by higher sensitivity for the other gut hormone. The aims of the present study were to investigate the bone microstructural morphology, as well as bone tissue properties, in double incretin receptor knock-out (DIRKO) mice. METHODS: Twenty-six-week-old DIRKO mice were age- and sex-matched with wild-type (WT) littermates. Bone microstructural morphology was assessed at the femur by microCT and quantitative X-ray imaging, while tissue properties were investigated by quantitative backscattered electron imaging and Fourier-transformed infrared microscopy. Bone mechanical response was assessed at the whole-bone- and tissue-level by 3-point bending and nanoindentation, respectively. RESULTS: As compared to WT animals, DIRKO mice presented significant augmentations in trabecular bone mass and trabecular number whereas bone outer diameter, cortical thickness, and cortical area were reduced. At the whole-bone-level, yield stress, ultimate stress, and post-yield work to fracture were significantly reduced in DIRKO animals. At the tissue-level, only collagen maturity was reduced by 9 % in DIRKO mice leading to reductions in maximum load, hardness, and dissipated energy. CONCLUSIONS: This study demonstrated the critical role of gut hormones in controlling bone microstructural morphology and tissue properties.


Subject(s)
Femur/pathology , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Adolescent , Animals , Biomechanical Phenomena/physiology , Bone Density/physiology , Femur/physiopathology , Gastric Inhibitory Polypeptide/deficiency , Gastric Inhibitory Polypeptide/genetics , Glucagon-Like Peptide 1/deficiency , Glucagon-Like Peptide 1/genetics , Glucose Intolerance/physiopathology , Glucose Tolerance Test/methods , Humans , Mice, Knockout , Stress, Mechanical , X-Ray Microtomography/methods
2.
Diabetes Obes Metab ; 16 Suppl 1: 87-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200301

ABSTRACT

Intracellular glucose signalling pathways control the secretion of glucagon and insulin by pancreatic islet α- and ß-cells, respectively. However, glucose also indirectly controls the secretion of these hormones through regulation of the autonomic nervous system that richly innervates this endocrine organ. Both parasympathetic and sympathetic nervous systems also impact endocrine pancreas postnatal development and plasticity in adult animals. Defects in these autonomic regulations impair ß-cell mass expansion during the weaning period and ß-cell mass adaptation in adult life. Both branches of the autonomic nervous system also regulate glucagon secretion. In type 2 diabetes, impaired glucose-dependent autonomic activity causes the loss of cephalic and first phases of insulin secretion, and impaired suppression of glucagon secretion in the postabsorptive phase; in diabetic patients treated with insulin, it causes a progressive failure of hypoglycaemia to trigger the secretion of glucagon and other counterregulatory hormones. Therefore, identification of the glucose-sensing cells that control the autonomic innervation of the endocrine pancreatic and insulin and glucagon secretion is an important goal of research. This is required for a better understanding of the physiological control of glucose homeostasis and its deregulation in diabetes. This review will discuss recent advances in this field of investigation.


Subject(s)
Feedback, Physiological , Islets of Langerhans/innervation , Models, Biological , Neurons/physiology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Appetite Regulation , Cell Size , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Glucagon/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Glucose Transporter Type 2/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Nerve Tissue Proteins/metabolism , Neurons/pathology , Parasympathetic Nervous System/cytology , Parasympathetic Nervous System/pathology , Parasympathetic Nervous System/physiopathology , Solitary Nucleus/physiology , Solitary Nucleus/physiopathology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/pathology , Sympathetic Nervous System/physiopathology
3.
J Intern Med ; 274(3): 203-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23751050

ABSTRACT

In healthy individuals, insulin resistance is associated with physiological conditions such as pregnancy or body weight gain and triggers an increase in beta cell number and insulin secretion capacity to preserve normoglycaemia. Failure of this beta cell compensation capacity is a fundamental cause of diabetic hyperglycaemia. Incomplete understanding of the molecular mechanisms controlling the plasticity of adult beta cells mechanisms and how these cells fail during the pathogenesis of diabetes strongly limits the ability to develop new beta cell-specific therapies. Here, current knowledge of the signalling pathways controlling beta cell plasticity is reviewed, and possible directions for future research are discussed.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/therapy , Glucose/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Differentiation/physiology , Disease Progression , Female , Gastrointestinal Hormones/metabolism , Humans , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Hyperglycemia/therapy , Insulin Secretion , Male , Pregnancy , Risk Factors , Signal Transduction
4.
Nat Genet ; 17(3): 327-30, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9354799

ABSTRACT

Glut-2 is a low-affinity transporter present in the plasma membrane of pancreatic beta-cells, hepatocytes and intestine and kidney absorptive epithelial cells of mice. In beta-cells, Glut-2 has been proposed to be active in the control of glucose-stimulated insulin secretion (GSIS; ref. 2), and its expression is strongly reduced in glucose-unresponsive islets from different animal models of diabetes. However, recent investigations have yielded conflicting data on the possible role of Glut-2 in GSIS. Whereas some reports have supported a specific role for Glut-2 (refs 5,6), others have suggested that GSIS could proceed normally even in the presence of low or almost undetectable levels of this transporter. Here we show that homozygous, but not heterozygous, mice deficient in Glut-2 are hyperglycaemic and relatively hypo-insulinaemic and have elevated plasma levels of glucagon, free fatty acids and beta-hydroxybutyrate. In vivo, their glucose tolerance is abnormal. In vitro, beta-cells display loss of control of insulin gene expression by glucose and impaired GSIS with a loss of first phase but preserved second phase of secretion, while the secretory response to non-glucidic nutrients or to D-glyceraldehyde is normal. This is accompanied by alterations in the postnatal development of pancreatic islets, evidenced by an inversion of the alpha- to beta-cell ratio. Glut-2 is thus required to maintain normal glucose homeostasis and normal function and development of the endocrine pancreas. Its absence leads to symptoms characteristic of non-insulin-dependent diabetes mellitus.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Monosaccharide Transport Proteins/genetics , Nerve Tissue Proteins , Animals , Blood Glucose/analysis , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Glucagon/metabolism , Glucose/pharmacology , Glucose Transporter Type 1 , Glucose Transporter Type 2 , Glucose Transporter Type 3 , Homozygote , Hyperglycemia/genetics , Insulin Secretion , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monosaccharide Transport Proteins/metabolism
5.
Diabetes Obes Metab ; 13 Suppl 1: 82-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21824260

ABSTRACT

Glucose homeostasis requires the tight regulation of glucose utilization by liver, muscle and white or brown fat, and glucose production and release in the blood by liver. The major goal of maintaining glycemia at ∼ 5 mM is to ensure a sufficient flux of glucose to the brain, which depends mostly on this nutrient as a source of metabolic energy. This homeostatic process is controlled by hormones, mainly glucagon and insulin, and by autonomic nervous activities that control the metabolic state of liver, muscle and fat tissue but also the secretory activity of the endocrine pancreas. Activation or inhibition of the sympathetic or parasympathetic branches of the autonomic nervous systems are controlled by glucose-excited or glucose-inhibited neurons located at different anatomical sites, mainly in the brainstem and the hypothalamus. Activation of these neurons by hyper- or hypoglycemia represents a critical aspect of the control of glucose homeostasis, and loss of glucose sensing by these cells as well as by pancreatic ß-cells is a hallmark of type 2 diabetes. In this article, aspects of the brain-endocrine pancreas axis are reviewed, highlighting the importance of central glucose sensing in the control of counterregulation to hypoglycemia but also mentioning the role of the neural control in ß-cell mass and function. Overall, the conclusions of these studies is that impaired glucose homeostasis, such as associated with type 2 diabetes, but also defective counterregulation to hypoglycemia, may be caused by initial defects in glucose sensing.


Subject(s)
Autonomic Nervous System/metabolism , Blood Glucose/metabolism , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Glucose Transporter Type 2/metabolism , Insulin/metabolism , Autonomic Nervous System/physiology , Biological Transport , Diabetes Mellitus, Type 2/physiopathology , Homeostasis , Humans , Hyperglycemia/metabolism , Hypoglycemia/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Signal Transduction
7.
Diabetologia ; 52(10): 2159-68, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19644669

ABSTRACT

AIMS/HYPOTHESIS: High- vs low-glycaemic index (GI) diets unfavourably affect body fat mass and metabolic markers in rodents. Different effects of these diets could be age-dependent, as well as mediated, in part, by carbohydrate-induced stimulation of glucose-dependent insulinotrophic polypeptide (GIP) signalling. METHODS: Young-adult (16 weeks) and aged (44 weeks) male wild-type (C57BL/6J) and GIP-receptor knockout (Gipr ( -/- )) mice were exposed to otherwise identical high-carbohydrate diets differing only in GI (20-26 weeks of intervention, n = 8-10 per group). Diet-induced changes in body fat distribution, liver fat, locomotor activity, markers of insulin sensitivity and substrate oxidation were investigated, as well as changes in the gene expression of anorexigenic and orexigenic hypothalamic factors related to food intake. RESULTS: Body weight significantly increased in young-adult high- vs low-GI fed mice (two-way ANOVA, p < 0.001), regardless of the Gipr genotype. The high-GI diet in young-adult mice also led to significantly increased fat mass and changes in metabolic markers that indicate reduced insulin sensitivity. Even though body fat mass also slightly increased in high- vs low-GI fed aged wild-type mice (p < 0.05), there were no significant changes in body weight and estimated insulin sensitivity in these animals. However, aged Gipr ( -/- ) vs wild-type mice on high-GI diet showed significantly lower cumulative net energy intake, increased locomotor activity and improved markers of insulin sensitivity. CONCLUSIONS/INTERPRETATION: The metabolic benefits of a low-GI diet appear to be more pronounced in younger animals, regardless of the Gipr genotype. Inactivation of GIP signalling in aged animals on a high-GI diet, however, could be beneficial.


Subject(s)
Diet , Gastric Inhibitory Polypeptide/physiology , Glycemic Index , Age Factors , Animals , Blood Glucose/analysis , Body Composition , Calorimetry , Energy Intake/physiology , Glucose Tolerance Test , Insulin/blood , Male , Mice , Mice, Knockout , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
8.
J Cell Biol ; 123(6 Pt 2): 1687-94, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8276890

ABSTRACT

The biosynthesis, intracellular transport, and surface expression of the beta cell glucose transporter GLUT2 was investigated in isolated islets and insulinoma cells. Using a trypsin sensitivity assay to measure cell surface expression, we determined that: (a) greater than 95% of GLUT2 was expressed on the plasma membrane; (b) GLUT2 did not recycle in intracellular vesicles; and (c) after trypsin treatment, reexpression of the intact transporter occurred with a t1/2 of approximately 7 h. Kinetics of intracellular transport of GLUT2 was investigated in pulse-labeling experiments combined with glycosidase treatment and the trypsin sensitivity assay. We determined that transport from the endoplasmic reticulum to the trans-Golgi network (TGN) occurred with a t1/2 of 15 min and that transport from the TGN to the plasma membrane required a similar half-time. When added at the start of a pulse-labeling experiment, brefeldin A prevented exit of GLUT2 from the endoplasmic reticulum. When the transporter was first accumulated in the TGN during a 15-min period of chase, but not following a low temperature (22 degrees C) incubation, addition of brefeldin A (BFA) prevented subsequent surface expression of the transporter. This indicated that brefeldin A prevented GLUT2 exit from the TGN by acting at a site proximal to the 22 degrees C block. Together, these data demonstrate that GLUT2 surface expression in beta cells is via the constitutive pathway, that transport can be blocked by BFA at two distinct steps and that once on the surface, GLUT2 does not recycle in intracellular vesicles.


Subject(s)
Cyclopentanes/pharmacology , Golgi Apparatus/metabolism , Insulinoma/metabolism , Islets of Langerhans/metabolism , Monosaccharide Transport Proteins/biosynthesis , Pancreatic Neoplasms/metabolism , Protein Synthesis Inhibitors/pharmacology , Animals , Blotting, Western , Brefeldin A , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Glucose Transporter Type 2 , Golgi Apparatus/drug effects , Kinetics , Monosaccharide Transport Proteins/analysis , Monosaccharide Transport Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Protein Structure, Secondary , Rats , Rats, Sprague-Dawley , Trypsin/pharmacology , Tumor Cells, Cultured
9.
J Cell Biol ; 94(1): 115-22, 1982 Jul.
Article in English | MEDLINE | ID: mdl-6981649

ABSTRACT

The vitamin D-dependent calcium binding protein (CaBP) of chick duodenum has been localized by immunocytochemistry and by radioimmunoassay. Light microscopically, CaBP was seen to be present in the absorptive cells of the villi while in other cell types of the villi and the crypts, including goblet cells and endocrine cells, no CaBP was seen. At the electron microscopic level, CaBP was shown to be localized in the cytosol and the euchromatin of the nucleus but not in membrane-bounded cytoplasmic compartments. Quantitative evaluation of the immunocytochemical protein A-gold label showed that the terminal web and the cytosol of basal cellular regions were most highly labeled while the brush border was weakly labeled. The radioimmunoassay evaluation of intestinal subcellular fractions indicated that 96% of the homogenate CaBP is in the cytosol high-speed supernatant fraction. Collectively, these results support the hypothesis that the vitamin D-dependent intestinal CaBP may play a role in either regulation of intracellular calcium concentration or movement of calcium across the brush border membrane from the gut lumen.


Subject(s)
Duodenum/metabolism , Ergocalciferols/metabolism , Animals , Calcium/metabolism , Cell Fractionation , Cell Nucleus/metabolism , Chickens , Cytoplasm/metabolism , Duodenum/ultrastructure , Epithelium/metabolism , In Vitro Techniques , Microvilli/metabolism , Organoids/metabolism
10.
Science ; 245(4915): 295-7, 1989 Jul 21.
Article in English | MEDLINE | ID: mdl-2665080

ABSTRACT

Immunocytochemical techniques revealed that the "liver-type" glucose transporter is present in the insulin-producing beta cells of rat pancreatic islets but not in other islet endocrine cells. Ultrastructural analysis of the transporter by the protein A-gold technique showed that it is restricted to certain domains of the plasma membrane, its density being sixfold higher in microvilli facing adjacent endocrine cells than in the flat regions of the plasma membrane. These results support a possible role for this glucose transporter in glucose sensing by beta cells and provide evidence that these cells are polarized.


Subject(s)
Islets of Langerhans/analysis , Monosaccharide Transport Proteins/analysis , Cell Membrane/analysis , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Islets of Langerhans/ultrastructure , Microscopy, Electron
11.
Science ; 214(4517): 197-200, 1981 Oct 09.
Article in English | MEDLINE | ID: mdl-7025212

ABSTRACT

A vitamin D--dependent calcium binding protein in the chick kidney that was detected by immunocytochemical techniques was localized exclusively in the distal convoluted tubule, the initial collecting tubule, and the early part of the collecting tubule. The intercalated (mitochondria-rich) cells in these tubular segments were negative for the calcium binding protein. Subcellularly, the protein was found in the cytosol and the nucleus of the tubular cells. The results suggest a role for vitamin D--dependent calcium binding protein in intracellular calcium metabolism rather than a direct involvement in membrane-mediated calcium reabsorption in the avian kidney.


Subject(s)
Calcium-Binding Proteins/metabolism , Kidney/metabolism , S100 Calcium Binding Protein G/metabolism , Animals , Cell Nucleus/metabolism , Chickens , Cytoplasm/metabolism , Immunoenzyme Techniques , Kidney/ultrastructure , Kidney Tubules/metabolism , Male , S100 Calcium Binding Protein G/immunology
13.
Int J Obes (Lond) ; 32 Suppl 6: S62-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19079282

ABSTRACT

The control of body weight and of blood glucose concentrations depends on the exquisite coordination of the function of several organs and tissues, in particular the liver, muscle and fat. These organs and tissues have major roles in the use and storage of nutrients in the form of glycogen or triglycerides and in the release of glucose or free fatty acids into the blood, in periods of metabolic needs. These mechanisms are tightly regulated by hormonal and nervous signals, which are generated by specialized cells that detect variations in blood glucose or lipid concentrations. The hormones insulin and glucagon not only regulate glycemic levels through their action on these organs and the sympathetic and parasympathetic branches of the autonomic nervous system, which are activated by glucose or lipid sensors, but also modulate pancreatic hormone secretion and liver, muscle and fat glucose and lipid metabolism. Other signaling molecules, such as the adipocyte hormones leptin and adiponectin, have circulating plasma concentrations that reflect the level of fat stored in adipocytes. These signals are integrated at the level of the hypothalamus by the melanocortin pathway, which produces orexigenic and anorexigenic neuropeptides to control feeding behavior, energy expenditure and glucose homeostasis. Work from several laboratories, including ours, has explored the physiological role of glucose as a signal that regulates these homeostatic processes and has tested the hypothesis that the mechanism of glucose sensing that controls insulin secretion by the pancreatic beta-cells is also used by other cell types. I discuss here evidence for these mechanisms, how they integrate signals from other nutrients such as lipids and how their deregulation may initiate metabolic diseases.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/etiology , Glucagon/metabolism , Insulin/metabolism , Obesity/etiology , Animals , Appetite Regulation/physiology , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acids/metabolism , Mice , Obesity/metabolism , Pancreas/metabolism , Portal System/metabolism , Rats
15.
J Clin Invest ; 86(3): 986-92, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2203828

ABSTRACT

The "erythroid/brain" glucose transporter (GT) isoform is expressed only in a subset of hepatocytes, those forming the first row around the terminal hepatic venules, while the "liver" GT is expressed in all hepatocytes. After 3 d of starvation, a three- to fourfold elevation of expression of the erythroid/brain GT mRNA and protein is detected in the liver as a whole; this correlates with the expression of this GT in more hepatocytes, those forming the first three to four rows around the hepatic venules. Starvation-dependent expression of the erythroid/brain GT on the plasma membrane of these additional hepatocytes is lost within 3 h of glucose refeeding; however, by immunoblotting we show that the protein is still present. Its loss from the surface is possibly explained by internalization.


Subject(s)
Liver/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Membrane/metabolism , Fluorescent Antibody Technique , Gene Expression , Glucose/pharmacology , Monosaccharide Transport Proteins/classification , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Starvation , Time Factors , Tissue Distribution
16.
J Clin Invest ; 90(1): 77-85, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1634622

ABSTRACT

Glucose-induced insulin secretion by beta cells of diabetic db/db mice was studied by a pancreas perfusion technique, and the levels of GLUT2 protein in pancreatic islets were assessed by immunofluorescence microscopy and protein blot analysis. Beta cells from diabetic mice had a high basal rate of insulin secretion; they did not respond to glucose stimulation but displayed a normal secretory response to arginine. At the same time, GLUT2 expression by db/db islets was lost whereas beta cells from nondiabetic db/+ mice expressed high levels of this transporter. GLUT2 levels in liver or kidney of diabetic mice were, however, mostly unaltered. Transplanting islets from db/db mice under the kidney capsule of db/+ mice restored normal GLUT2 levels. Conversely, transplantation of db/+ islets into db/db mice induced the disappearance of GLUT2 expression. When islets from db/+ mice were transplanted under the kidney capsule of streptozocin-diabetic mice, the immunodetection of GLUT2 also disappeared. We conclude that: (a) GLUT2 expression is decreased in glucose-unresponsive beta cells from db/db mice; (b) the decreased expression of GLUT2 is reversible; (c) the loss of GLUT2 expression is induced by the diabetic environment of db/db and streptozocin-induced diabetic mice. These observations together with previously published data suggest that a factor different from glucose or insulin regulates the beta cell expression of GLUT2.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/pharmacology , Islets of Langerhans/metabolism , Monosaccharide Transport Proteins/analysis , Animals , Hyperglycemia/metabolism , Insulin/blood , Insulin/metabolism , Insulin Secretion , Mice , Perfusion
17.
J Clin Invest ; 88(5): 1553-8, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1939644

ABSTRACT

It has been proposed that the mercurial-sensitive water transporter in mammalian erythrocytes is the anion exchanger band 3 (AE1) and/or the glucose transporter, band 4.5 (GLUT1). Using a functional assay for water channel expression in Xenopus oocytes (Zhang, R., K. A. Logee, and A. S. Verkman. 1990. J. Biol. Chem. 265:15375-15378), we compared osmotic water permeability (Pf) of oocytes injected with water, reticulocyte mRNA, AE1 mRNA, and GLUT1 mRNA. Injection of oocytes with 5-50 ng of in vitro-transcribed AE1 mRNA had no effect on Pf, but increased trans-stimulated 36Cl uptake greater than fourfold in a dinitro-disulfonic stilbene (DNDS)-inhibitable manner. Injection with 1-50 ng of in vitro-transcribed GLUT1 mRNA increased 3H-methylglucose uptake greater than 15-fold in a cytochalasin B-sensitive manner and increased Pf from (3.7 +/- 0.4) x 10(-4) cm/s (SE, n = 16, 10 degrees C) in water-injected oocytes up to (13 +/- 1) x 10(-4) cm/s (n = 18). Both the increments in sugar and water transport were inhibited by cytochalasin B (25 microM) and phloretin (0.2 mM); neither was inhibited by 0.3 mM HgCl2. In oocytes injected with 50 ng of rabbit reticulocyte mRNA, the Pf of (18 +/- 2) x 10(-4) cm/s (n = 18) was reduced to (4.0 +/- 0.6) x 10(-4) cm/s (n = 10) by HgCl2, but was not inhibited by DNDS (0.4 mM), cytochalasin B or phloretin. Coinjection of reticulocyte mRNA with antisense oligodeoxyribonucleotides against AE1 or GLUT1 did not affect Pf, but inhibited completely the incremental uptake of 36Cl or 3H-methylglucose, respectively. Expression of size-fractionated mRNA from reticulocyte gave a 2-2.5-kb size for water channel mRNA, less than the 4-4.5-kb size for the Cl transporter. These results provide evidence that facilitated water transport in erythrocytes is mediated not by bands 3 or 4.5, but by distinct water transport protein(s).


Subject(s)
Anion Exchange Protein 1, Erythrocyte/physiology , Body Water/metabolism , Erythrocytes/metabolism , Monosaccharide Transport Proteins/physiology , Oocytes/metabolism , Animals , Base Sequence , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/pharmacology , Rabbits , Xenopus laevis
18.
J Clin Invest ; 86(5): 1615-22, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2243134

ABSTRACT

Previous studies from our laboratories have suggested a defect in glucose transport in islets isolated from BB rats on the first day of overt diabetes. To quantitate by immunostaining the glucose transporter of beta-cells (GLUT-2) before and at the onset of autoimmune diabetes we employed an antibody to its COOH-terminal octapeptide. On the first day of overt diabetes, defined as the day the daily blood glucose first reached 200 mg/dl, the volume density ratio of GLUT-2-positive to insulin-positive beta-cells was only 0.48 +/- 0.06, compared to 0.91 +/- 0.02 in age-matched nondiabetic diabetes-resistant controls (P less than 0.001). In age-matched nondiabetic diabetes-prone rats, most of which would have become diabetic, the ratio was 0.85 +/- 0.02, also less than the controls (P less than 0.05). Protein A-gold labeling of GLUT-2 in beta-cells of day 1 diabetic rats revealed 2.17 +/- 0.16 gold particles per micrometer length of microvillar plasma membranes compared to 3.91 +/- 0.14 in controls (P less than 0.001) and 2.87 +/- 0.24 in the nondiabetic diabetes-prone rats (P less than 0.02). Reduction in GLUT-2 correlates temporally with and may contribute to the loss of glucose-stimulated insulin secretion that precedes profound beta-cell depletion of autoimmune diabetes.


Subject(s)
B-Lymphocytes/metabolism , Diabetes Mellitus, Experimental/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , B-Lymphocytes/ultrastructure , Fluorescent Antibody Technique , Glucose/metabolism , Glucose/pharmacology , Immunohistochemistry , Insulin/metabolism , Male , Microscopy, Electron , Rats , Rats, Inbred Strains
19.
Mol Cell Biol ; 12(1): 422-32, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1729614

ABSTRACT

High-level expression of the low-Km glucose transporter isoform GLUT-1 is characteristic of many cultured tumor and oncogene-transformed cells. In this study, we tested whether induction of GLUT-1 occurs in tumors in vivo. Normal mouse beta islet cells express the high-Km (approximately 20 mM) glucose transporter isoform GLUT-2 but not the low-Km (1 to 3 mM) GLUT-1. In contrast, a beta cell line derived from an insulinoma arising in a transgenic mouse harboring an insulin-promoted simian virus 40 T-antigen oncogene (beta TC3) expressed very low levels of GLUT-2 but high levels of GLUT-1. GLUT-1 protein was not detectable on the plasma membrane of islets or tumors of the transgenic mice but was induced in high amounts when the tumor-derived beta TC3 cells were grown in tissue culture. GLUT-1 expression in secondary tumors formed after injection of beta TC3 cells into mice was reduced. Thus, high-level expression of GLUT-1 in these tumor cells is characteristic of culture conditions and is not induced by the oncogenic transformation; indeed, overnight culture of normal pancreatic islets causes induction of GLUT-1. We also investigated the relationship between expression of the different glucose transporter isoforms by islet and tumor cells and induction of insulin secretion by glucose. Prehyperplastic transgenic islet cells that expressed normal levels of GLUT-2 and no detectable GLUT-1 exhibited an increased sensitivity to glucose, as evidenced by maximal insulin secretion at lower glucose concentrations, compared with that exhibited by normal islets. Further, hyperplastic islets and primary and secondary tumors expressed low levels of GLUT-2 and no detectable GLUT-1 on the plasma membrane; these cells exhibited high basal insulin secretion and responded poorly to an increase in extracellular glucose. Thus, abnormal glucose-induced secretion of insulin in prehyperplastic islets in mice was independent of changes in GLUT-2 expression and did not require induction of GLUT-1 expression.


Subject(s)
Antigens, Viral, Tumor/metabolism , Islets of Langerhans/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Transformation, Neoplastic , Cell Transformation, Viral , Cells, Cultured , Fluorescent Antibody Technique , Glucose/metabolism , Insulin/physiology , Islets of Langerhans/cytology , Mice , Mice, Inbred C57BL , Molecular Conformation , Monosaccharide Transport Proteins/chemistry
20.
Clin Nutr ; 36(2): 355-363, 2017 04.
Article in English | MEDLINE | ID: mdl-27686693

ABSTRACT

Growing evidence underscores the important role of glycemic control in health and recovery from illness. Carbohydrate ingestion in the diet or administration in nutritional support is mandatory, but carbohydrate intake can adversely affect major body organs and tissues if resulting plasma glucose becomes too high, too low, or highly variable. Plasma glucose control is especially important for patients with conditions such as diabetes or metabolic stress resulting from critical illness or surgery. These patients are particularly in need of glycemic management to help lessen glycemic variability and its negative health consequences when nutritional support is administered. Here we report on recent findings and emerging trends in the field based on an ESPEN workshop held in Venice, Italy, 8-9 November 2015. Evidence was discussed on pathophysiology, clinical impact, and nutritional recommendations for carbohydrate utilization and management in nutritional support. The main conclusions were: a) excess glucose and fructose availability may exacerbate metabolic complications in skeletal muscle, adipose tissue, and liver and can result in negative clinical impact; b) low-glycemic index and high-fiber diets, including specialty products for nutritional support, may provide metabolic and clinical benefits in individuals with obesity, insulin resistance, and diabetes; c) in acute conditions such as surgery and critical illness, insulin resistance and elevated circulating glucose levels have a negative impact on patient outcomes and should be prevented through nutritional and/or pharmacological intervention. In such acute settings, efforts should be implemented towards defining optimal plasma glucose targets, avoiding excessive plasma glucose variability, and optimizing glucose control relative to nutritional support.


Subject(s)
Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/adverse effects , Insulin Resistance , Nutrition Policy , Nutritional Support , Blood Glucose/metabolism , Carbohydrate Metabolism , Diet , Evidence-Based Medicine , Glycemic Index , Humans , Hyperglycemia/etiology , Hyperglycemia/therapy , Hypoglycemia/etiology , Hypoglycemia/therapy , Italy , Nutritional Requirements , Risk Factors , Societies, Scientific
SELECTION OF CITATIONS
SEARCH DETAIL