Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Pediatr Surg Int ; 32(3): 221-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26527582

ABSTRACT

PURPOSE: The gold standard for the diagnosis of Hirschsprung's disease (HSCR) is the pathologic evaluation of a rectal biopsy that demonstrates the absence of ganglion cells and nerve fibre hypertrophy. However, it has been frequently reported that hypertrophic nerves may not be present in some variants like long-segment HSCR, total colonic aganglionosis, premature and very young infants. The aim of this study was to determine this association. METHODS: We performed a retrospective review of the HSCR database at our tertiary care children's hospital from 2000 to 2013. In order to analyse the relationship between the diameter of the nerve fibres and the level of aganglionosis, we classified the patient sample into two groups-fibres ≤40 and >40 µm. The groups were statistically compared with P < 0.05 being significant. RESULTS: Rectal biopsies of 92 patients confirmed as HSCR with definitive operation performed at the same institution were reviewed. The mean nerve diameter was 50.1 µm (range 20-87.5 µm). Nerve fibre diameter ≤40 µm was predictive of transition zone above the sigmoid colon. A specificity of 77.3 % and a likelihood ratio of 2.03 supported this perception. No correlation was noted between nerve fibre diameter and gestational age at birth, birth weight or age at biopsy. CONCLUSION: The absence of nerve fibre hypertrophy in the presence of aganglionosis on rectal biopsy specimens is predictive of long-segment HSCR.


Subject(s)
Colon, Sigmoid/pathology , Hirschsprung Disease/diagnosis , Hirschsprung Disease/pathology , Nerve Fibers/pathology , Biopsy , Child , Child, Preschool , Female , Humans , Hypertrophy , Infant , Infant, Newborn , Male , Reproducibility of Results , Retrospective Studies
2.
Hum Vaccin Immunother ; 10(8): 2417-20, 2014.
Article in English | MEDLINE | ID: mdl-25424949

ABSTRACT

BACKGROUND: In recent years, the number of varicella cases in adults has significantly increased in Beijing. However, the effect of the vaccination on varicella-related characteristics among adults has not been studied. METHODS AND RESULTS: Using data from the Infectious Disease Reporting System and the Immunization Information System, we compared the epidemiology and disease severity in breakthrough and unvaccinated varicella cases in adolescents and adults (≥ 15 year-old) from 2008 to 2011 in Beijing's Fengtai district, China. The results showed that the age (P = 0.003),contact history (90% vs. 73%, P = 0.019) and outbreak cases (10% vs. 1%, P < 0.0001) were significantly differently distributed between the two groups and that both the incidence of moderate-to-severe cases (26% vs. 45%, P = 0.035, OR = 0.446) and varicella-associated fever (49% vs. 66%, P = 0.068, OR = 0.534) were either significantly lower or trended to be lower in the breakthrough group than in the unvaccinated group. Additionally,vaccine effectiveness against moderate-to-severe cases of varicella was 55.4%. CONCLUSION: Altogether, these results indicate that vaccination against varicella among adolescents and adults affected the epidemiology and attenuated the disease severity of the cases. The Results from this study will provide useful information for the prevention of varicella in adolescents and adults.


Subject(s)
Chickenpox Vaccine/administration & dosage , Chickenpox Vaccine/immunology , Chickenpox/pathology , Chickenpox/prevention & control , Adolescent , Adult , Chickenpox/epidemiology , Child , Child, Preschool , China/epidemiology , Female , Humans , Incidence , Male , Severity of Illness Index , Young Adult
3.
J Clin Invest ; 122(1): 388-402, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22182839

ABSTRACT

Glucagon-like peptide-1 (GLP-1) circulates at low levels and acts as an incretin hormone, potentiating glucose-dependent insulin secretion from islet ß cells. GLP-1 also modulates gastric emptying and engages neural circuits in the portal region and CNS that contribute to GLP-1 receptor-dependent (GLP-1R-dependent) regulation of glucose homeostasis. To elucidate the importance of pancreatic GLP-1R signaling for glucose homeostasis, we generated transgenic mice that expressed the human GLP-1R in islets and pancreatic ductal cells (Pdx1-hGLP1R:Glp1r-/- mice). Transgene expression restored GLP-1R-dependent stimulation of cAMP and Akt phosphorylation in isolated islets, conferred GLP-1R-dependent stimulation of ß cell proliferation, and was sufficient for restoration of GLP-1-stimulated insulin secretion in perifused islets. Systemic GLP-1R activation with the GLP-1R agonist exendin-4 had no effect on food intake, hindbrain c-fos expression, or gastric emptying but improved glucose tolerance and stimulated insulin secretion in Pdx1-hGLP1R:Glp1r-/- mice. i.c.v. GLP-1R blockade with the antagonist exendin(9-39) impaired glucose tolerance in WT mice but had no effect in Pdx1-hGLP1R:Glp1r-/- mice. Nevertheless, transgenic expression of the pancreatic GLP-1R was sufficient to normalize both oral and i.p. glucose tolerance in Glp1r-/- mice. These findings illustrate that low levels of endogenous GLP-1 secreted from gut endocrine cells are capable of augmenting glucoregulatory activity via pancreatic GLP-1Rs independent of communication with neural pathways.


Subject(s)
Glucose/metabolism , Incretins/metabolism , Pancreas/physiology , Receptors, Glucagon/metabolism , Animals , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Homeostasis , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurosecretory Systems/physiology , Peptide Fragments/pharmacology , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
4.
Am J Physiol Endocrinol Metab ; 296(6): E1251-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19223654

ABSTRACT

Thioredoxin-interacting protein (TxNIP) is an endogenous inhibitor of thioredoxin, a ubiquitous thiol oxidoreductase, that regulates cellular redox status. Diabetic mice exhibit increased expression of TxNIP in pancreatic islets, and recent studies suggest that TxNIP is a proapoptotic factor in beta-cells that may contribute to the development of diabetes. Here, we examined the role of TxNIP deficiency in vivo in the development of insulin-deficient diabetes and whether it impacted on pancreatic beta-cell mass and/or insulin secretion. TxNIP-deficient (Hcb-19/TxNIP(-/-)) mice had lower baseline glycemia, higher circulating insulin concentrations, and higher total pancreatic insulin content and beta-cell mass than control mice (C3H). Hcb-19/TxNIP(-/-) did not develop hyperglycemia when injected with standard multiple low doses of streptozotocin (STZ), in contrast to C3H controls. Surprisingly, although beta-cell mass remained higher in Hcb-19/TxNIP(-/-) mice compared with C3H after STZ exposure, the relative decrease induced by STZ was as great or even greater in the TxNIP-deficient animals. Consistently, cultured pancreatic INS-1 cells transfected with small-interfering RNA against TxNIP were more sensitive to cell death induced by direct exposure to STZ or to the combination of inflammatory cytokines interleukin-1beta, interferon-gamma, and tumor necrosis factor-alpha. Furthermore, when corrected for insulin content, isolated pancreatic islets from TxNIP(-/-) mice exhibited reduced glucose-induced insulin secretion. These data indicate that TxNIP functions as a regulator of beta-cell mass and influences insulin secretion. In conclusion, the relative resistance of TxNIP-deficient mice to STZ-induced diabetes appears to be because of an increase in beta-cell mass. However, TxNIP deficiency is associated with sensitization to STZ- and cytokine-induced beta-cell death, indicating complex regulatory roles of TxNIP under different physiological and pathological conditions.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/cytology , Thioredoxins/genetics , Thioredoxins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Cell Cycle Proteins , Cell Line , Cytokines/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Dose-Response Relationship, Drug , Glucose/pharmacology , In Situ Nick-End Labeling , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Rats , Streptozocin/pharmacology
5.
Ann N Y Acad Sci ; 1152: 154-64, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19161386

ABSTRACT

Neuroendocrine pancreatic islet beta-cells secrete the hormone insulin in response to glucose stimulation and adapt efficiently to increased demand by peripheral tissues to maintain glucose homeostasis. Insulin is packed within dense-core granules, which traffic and dock onto the plasma membrane whereby a Ca(2+) stimulus evokes exocytosis by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE), complex-mediated, membrane fusion. Recent studies have unveiled postdocking steps mediated by "priming" factors that influence SNARE complex assembly to confer fusion readiness to the docked granules. This review will summarize recent insights into the priming role for Munc13 in the exocytosis of insulin granules. We present evidence for the interaction of Munc13-1 with exocytotic substrates involved in cAMP-mediated potentiation of insulin release, the latter we show to mediate enhanced granule-to-granule fusion events underlying compound exocytosis. We thus also further review the current understanding of granule-to-granule fusion. As agents acting on cAMP signaling are clinically used to augment insulin release in diabetes, this better understanding of priming steps may reveal additional novel therapeutic strategies to increase the capacity for insulin release to improve the treatment of diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Exocytosis , Insulin/metabolism , Animals , Calcium/metabolism , Humans , Insulin Secretion , Intracellular Membranes/metabolism , Nerve Tissue Proteins/metabolism
6.
J Biol Chem ; 283(26): 17939-45, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18426794

ABSTRACT

RalA is a small GTPase that is thought to facilitate exocytosis through its direct interaction with the mammalian exocyst complex. In this study, we report an essential role for RalA in regulated insulin secretion from pancreatic beta cells. We employed lentiviral-mediated delivery of RalA short hairpin RNAs to deplete endogenous RalA protein in mouse pancreatic islets and INS-1 beta cells. Perifusion of mouse islets depleted of RalA protein exhibited inhibition of both first and second phases of glucose-stimulated insulin secretion. Consistently, INS-1 cells depleted of RalA caused a severe inhibition of depolarization-induced insulin exocytosis determined by membrane capacitance, including a reduction in the size of the ready-releasable pool of insulin granules and a reduction in the subsequent mobilization and exocytosis of the reserve pool of granules. Collectively, these data suggest that RalA is a critical component in biphasic insulin release from pancreatic beta cells.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/cytology , Insulin/metabolism , ral GTP-Binding Proteins/physiology , Animals , Apoptosis , Exocytosis , Humans , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Rats
7.
Mol Biol Cell ; 19(2): 722-34, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077557

ABSTRACT

Although Munc18-1 was originally identified as a syntaxin1-interacting protein, the physiological significance of this interaction remains unclear. In fact, recent studies of Munc18-1 mutants have suggested that Munc18-1 plays a critical role for docking of secretory vesicles, independent of syntaxin1 regulation. Here we investigated the role of Munc18-1 in syntaxin1 localization by generating stable neuroendocrine cell lines in which Munc18-1 was strongly down-regulated. In these cells, the secretion capability, as well as the docking of dense-core vesicles, was significantly reduced. More importantly, not only was the expression level of syntaxin1 reduced, but the localization of syntaxin1 at the plasma membrane was also severely perturbed. The mislocalized syntaxin1 resided primarily in the perinuclear region of the cells, in which it was highly colocalized with Secretogranin II, a marker protein for dense-core vesicles. In contrast, the expression level and the plasma membrane localization of SNAP-25 were not affected. Furthermore, the syntaxin1 localization and the secretion capability were restored upon transfection-mediated reintroduction of Munc18-1. Our results indicate that endogenous Munc18-1 plays a critical role for the plasma membrane localization of syntaxin1 in neuroendocrine cells and therefore necessitates the interpretation of Munc18-1 mutant phenotypes to be in terms of mislocalized syntaxin1.


Subject(s)
Cell Membrane/metabolism , Munc18 Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Animals , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Humans , Microscopy, Confocal , Munc18 Proteins/genetics , Nerve Growth Factor/pharmacology , PC12 Cells , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Secretory Vesicles/drug effects , Secretory Vesicles/ultrastructure , Subcellular Fractions/metabolism , Transfection
8.
Endocr Rev ; 28(6): 653-63, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17878408

ABSTRACT

The three SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins, syntaxin, SNAP25 (synaptosome-associated protein of 25 kDa), and synaptobrevin, constitute the minimal machinery for exocytosis in secretory cells such as neurons and neuroendocrine cells by forming a series of complexes prior to and during vesicle fusion. It was subsequently found that these SNARE proteins not only participate in vesicle fusion, but also tether with voltage-dependent Ca(2+) channels to form an excitosome that precisely regulates calcium entry at the site of exocytosis. In pancreatic islet beta-cells, ATP-sensitive K(+) (K(ATP)) channel closure by high ATP concentration leads to membrane depolarization, voltage-dependent Ca(2+) channel opening, and insulin secretion, whereas subsequent opening of voltage-gated K(+) (Kv) channels repolarizes the cell to terminate exocytosis. We have obtained evidence that syntaxin-1A physically interacts with Kv2.1 (the predominant Kv in beta-cells) and the sulfonylurea receptor subunit of beta-cell K(ATP) channel to modify their gating behaviors. A model has proposed that the conformational changes of syntaxin-1A during exocytosis induce distinct functional modulations of K(ATP) and Kv2.1 channels in a manner that optimally regulates cell excitability and insulin secretion. Other proteins involved in exocytosis, such as Munc-13, tomosyn, rab3a-interacting molecule, and guanyl nucleotide exchange factor II, have also been implicated in direct or indirect regulation of beta-cell ion channel activities and excitability. This review discusses this interesting aspect that exocytotic proteins not only promote secretion per se, but also fine-tune beta-cell excitability via modulation of ion channel gating.


Subject(s)
Insulin-Secreting Cells/metabolism , Ion Channel Gating/physiology , KATP Channels/metabolism , Potassium Channels, Voltage-Gated/metabolism , SNARE Proteins/physiology , Syntaxin 1/physiology , Animals , Exocytosis/physiology , Humans
9.
Pancreas ; 35(3): e45-54, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17895835

ABSTRACT

OBJECTIVES: Using FM1-43 epifluorescence imaging and electron microscopy, we recently reported that glucagon-like peptide (GLP-1)-mediated cyclic adenosine monophosphate (cAMP) potentiation of insulin secretion markedly promotes the number of plasma membrane (PM) exocytic sites and insulin secretory granule (SG)-to-granule fusions underlying compound and sequential exocytosis. METHODS: Here, we used FM1-43 imaging to dissect the distinct contributions of putative GLP-1/cAMP activated substrates--exchange protein directly activated by cAMP (EPAC) and protein kinase A (PKA)--in mediating these exocytic events. RESULTS: Like GLP-1, cAMP activation by forskolin increased the number of PM exocytic sites (2.3-fold), which were mainly of the robust-sustained (55.8%) and stepwise-multiphasic (37.7%) patterns corresponding to compound and sequential SG-SG exocytosis, respectively, with few monophasic hotspots (6.5%) corresponding to single-granule exocytosis. Direct activation of EPAC by 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cAMP also increased the number of exocytic sites, but which were mainly multiphasic (60%) and monophasic (40%) hotspots. Protein kinase A inhibition by H89 blocked forskolin-evoked robust-sustained hotspots, while retaining multiphasic (47%) and monophasic (53%) hotspots. Consistently, PKA activation (N6-benzoyladenosine-3',5'-cAMP) evoked only multiphasic (60%) and monophasic (40%) hotspots. These results suggested that PKA activation is required but alone is insufficient to promote compound SG-SG fusions. 8-(4-Chloro-phenylthio)-2'-O-methyladenosine-3',5'-cAMP plus N6-benzoyladenosine-3',5'-cAMP stimulation completely reconstituted the effects of forskolin, including increasing the number of exocytic sites, with a similar pattern of robust-sustained (42.6%) and stepwise (39.6%) hotspots and few monophasic (17.8%) hotspots. CONCLUSIONS: The EPAC and PKA modulate both distinct and common exocytic steps to potentiate insulin exocytosis where (a) EPAC activation mobilizes SGs to fuse at the PM, thereby increasing number of PM exocytic sites; and (b) PKA and EPAC activation synergistically modulate SG-SG fusions underlying compound and sequential exocytoses.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Exocytosis/physiology , Guanine Nucleotide Exchange Factors/physiology , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Glucagon-Like Peptide 1/physiology , Insulin Secretion , Isoquinolines/pharmacology , Male , Membrane Fusion/physiology , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Secretory Vesicles/physiology , Sulfonamides/pharmacology
10.
Diabetes ; 56(10): 2579-88, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17639022

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) rescues insulin secretory deficiency in type 2 diabetes partly via cAMP actions on exchange protein directly activated by cAMP (Epac2) and protein kinase A (PKA)-activated Rab3A-interacting molecule 2 (Rim2). We had reported that haplodeficient Munc13-1(+/-) mouse islet beta-cells exhibited reduced insulin secretion, causing glucose intolerance. Munc13-1 binds Epac2 and Rim2, but their functional interactions remain unclear. RESEARCH DESIGN AND METHODS: We used Munc13-1(+/-) islet beta-cells to examine the functional interactions between Munc13-1 and Epac2 and PKA. GLP-1 stimulation of Munc13-1(+/-) islets normalized the reduced biphasic insulin secretion by its actions on intact islet cAMP production and normal Epac2 and Rim2 levels. RESULTS: To determine which exocytotic steps caused by Munc13-1 deficiency are rescued by Epac2 and PKA, we used patch-clamp capacitance measurements, showing that 1) cAMP restored the reduced readily releasable pool (RRP) and partially restored refilling of a releasable pool of vesicles in Munc13-1(+/-) beta-cells, 2) Epac-selective agonist [8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate] partially restored the reduced RRP and refilling of a releasable pool of vesicles, and 3) PKA blockade by H89 (leaving Epac intact) impaired cAMP ability to restore the RRP and refilling of a releasable pool of vesicles. Conversely, PKA-selective agonist (N(6)-benzoyladenosine-cAMP) completely restored RRP and partially restored refilling of a releasable pool of vesicles. To determine specific contributions within Epac-Rim2-Munc13-1 interaction sites accounting for cAMP rescue of exocytosis caused by Munc13-1 deficiency, we found that blockade of Rim2-Munc13-1 interaction with Rim-Munc13-1-binding domain peptide abolished cAMP rescue, whereas blockade of Epac-Rim2 interaction with Rim2-PDZ peptide only moderately reduced refilling with little effect on RRP. CONCLUSIONS: cAMP rescue of priming defects caused by Munc13-1 deficiency via Epac and PKA signaling pathways requires downstream Munc13-1-Rim2 interaction.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Exocytosis/physiology , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Nerve Tissue Proteins/physiology , Animals , Cyclic AMP/metabolism , Exocytosis/drug effects , Insulin/deficiency , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques
11.
Diabetes ; 56(9): 2302-11, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17563067

ABSTRACT

Inadequate pancreatic beta-cell mass resulting from excessive beta-cell apoptosis is a key defect in type 1 and type 2 diabetes. Caspases are the major molecules involved in apoptosis; however, in vivo roles of specific caspases in diabetes are unclear. The purpose of this study is to examine the role of Caspase (Casp)8 in beta-cells in vivo. Using the Cre-loxP system, mice lacking Casp8 in beta-cells (RIPcre(+)Casp8(fl/fl) mice) were generated to address the role of Casp8 in beta-cells in physiological and diabetes models. We show that islets isolated from RIPcre(+)Casp8(fl/fl) mice were protected from Fas ligand (FasL)-and ceramide-induced cell death. Furthermore, RIPcre(+)Casp8(fl/fl) mice were protected from in vivo models of type 1 and type 2 diabetes. In addition to being the central mediator of apoptosis in diabetes models, we show that Casp8 is critical for maintenance of beta-cell mass under physiological conditions. With aging, RIPcre(+)Casp8(fl/fl) mice gradually develop hyperglycemia and a concomitant decline in beta-cell mass. Their islets display decreased expression of molecules involved in insulin/IGF-I signaling and show decreased pancreatic duodenal homeobox-1 and cAMP response element binding protein expression. At the level of individual islets, we observed increased insulin secretory capacity associated with increased expression of exocytotic proteins. Our results show distinct context-specific roles of Casp8 in physiological and disease states; Casp8 is essential for beta-cell apoptosis in type 1 and type 2 diabetes models and in regulating beta-cell mass and insulin secretion under physiological conditions.


Subject(s)
Caspase 8/genetics , Caspase 8/metabolism , Diabetes Mellitus, Experimental/enzymology , Insulin-Secreting Cells/enzymology , Animals , Blood Glucose/metabolism , Cell Death , Cell Separation , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
12.
Diabetes ; 55(5): 1421-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16644700

ABSTRACT

Munc13-1 is a diacylglycerol (DAG) receptor that is essential for synaptic vesicle priming. We recently showed that Munc13-1 is expressed in rodent and human islet beta-cells and that its levels are reduced in islets of type 2 diabetic humans and rat models, suggesting that Munc13-1 deficiency contributes to the abnormal insulin secretion in diabetes. To unequivocally demonstrate the role of Munc13-1 in insulin secretion, we studied heterozygous Munc13-1 knockout mice (+/-), which exhibited elevated glucose levels during intraperitoneal glucose tolerance tests with corresponding lower serum insulin levels. Munc13-1(+/-) mice exhibited normal insulin tolerance, indicating that a primary islet beta-cell secretory defect is the major cause of their hyperglycemia. Consistently, glucose-stimulated insulin secretion was reduced 50% in isolated Munc13-1(+/-) islets and was only partially rescued by phorbol ester potentiation. The corresponding alterations were minor in mice expressing one allele of a Munc13-1 mutant variant, which does not bind DAG (H567K/+). Capacitance measurements of Munc13-1(+/-) and Munc13-1(H567k/+) islet beta-cells revealed defects in granule priming, including the initial size and refilling of the releasable pools, which become accentuated by phorbol ester potentiation. We conclude that Munc13-1 plays an important role in glucose-stimulated insulin secretion and that Munc13-1 deficiency in the pancreatic islets as occurs in diabetes can reduce insulin secretion sufficient to cause abnormal glucose homeostasis.


Subject(s)
Glucose Intolerance/genetics , Insulin/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Animals , Brain/metabolism , Brain/physiology , Crosses, Genetic , Glucose Tolerance Test , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques , Polymerase Chain Reaction
13.
Diabetes ; 54(9): 2734-43, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123364

ABSTRACT

Glucagon-like peptide 1 (GLP-1) has been postulated to potentiate insulin secretion by cAMP-mediated enhancement of mobilization and priming of secretory granules, but the precise exocytic events are unknown. We used epi-fluorescent microscopy of the fluorescent dye FM1-43, which incorporates into the plasma membrane and the exocytosing secretory granules (appearing as plasma membrane hotspots). KCl evoked exocytosis of 1.8 +/- 0.5 hotspots/rat beta-cell at the cell periphery, 82% of which are single transient increases of low amplitudes (151 +/- 7%), suggesting single secretory granule exocytosis; and the remaining 18% are stepwise increases in plasma membrane hotspots with higher amplitudes (170 +/- 9%), suggesting sequential secretory granule to secretory granule exocytic fusions. Addition of GLP-1 increased the hotspots to 6.0 +/- 0.7/beta-cell and exhibited a larger number of stepwise (41%) than transient (10%) increases with higher amplitudes of 259 +/- 19 and 278 +/- 23%, respectively. More interestingly, GLP-1 also evoked a robust and sustained pattern (49%) with even higher amplitudes of 354 +/- 18%, which are likely accelerated sequential secretory granule-secretory granule fusions. Electron microscopy studies collaborated with these imaging results, showing that GLP-1 increased the number of docked secretory granules at the plasma membrane and also increased the number of events showing direct contact of oncoming secretory granules with secretory granules undergoing exocytosis. We conclude that the potentiation of insulin secretion by GLP-1 is contributed by the mobilization of more insulin secretory granules to dock at the plasma membrane and the acceleration of sequential secretory granule-secretory granule fusions.


Subject(s)
Exocytosis/physiology , Islets of Langerhans/physiology , Secretory Vesicles/physiology , Animals , Cells, Cultured , Insulin/metabolism , Islets of Langerhans/ultrastructure , Male , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 279(23): 24685-91, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15073181

ABSTRACT

In pancreatic beta-cells, the predominant voltage-gated Ca(2+) channel (Ca(V)1.2) and K(+) channel (K(V)2.1) are directly coupled to SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor) proteins. These SNARE proteins modulate channel expression and gating and closely associate these channels with the insulin secretory vesicles. We show that K(V)2.1 and Ca(V)1.2, but not K(V)1.4, SUR1, or Kir6.2, target to specialized cholesterol-rich lipid raft domains on beta-cell plasma membranes. Similarly, the SNARE proteins syntaxin 1A, SNAP-25, and VAMP-2, but not Munc-13-1 or n-Sec1, are associated with lipid rafts. Disruption of the lipid rafts by depleting membrane cholesterol with methyl-beta-cyclodextrin shunts K(V)2.1, Ca(V)1.2, and SNARE proteins out of lipid rafts. Furthermore, methyl-beta-cyclodextrin inhibits K(V)2.1 but not Ca(V)1.2 channel activity and enhances single-cell exocytic events and insulin secretion. Membrane compartmentalization of ion channels and SNARE proteins in lipid rafts may be critical for the temporal and spatial coordination of insulin release, forming what has been described as the excitosome complex.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Membrane Microdomains/chemistry , Potassium Channels, Voltage-Gated , Potassium Channels/chemistry , Vesicular Transport Proteins , beta-Cyclodextrins , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Cholesterol/chemistry , Cholesterol/metabolism , Cricetinae , Cyclodextrins/chemistry , Delayed Rectifier Potassium Channels , Electrophysiology , Exocytosis , Insulin Secretion , Ions , Membrane Proteins/metabolism , Microscopy, Confocal , Potassium/metabolism , Protein Structure, Tertiary , Rats , SNARE Proteins , Shab Potassium Channels , Syntaxin 1
15.
Am J Pathol ; 164(1): 253-62, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14695338

ABSTRACT

S100A4/Mts1 confers a metastatic phenotype in tumor cells and may also be related to resistance to apoptosis and angiogenesis. Approximately 5% of transgenic mice overexpressing S100A4/Mts1 develop pulmonary arterial changes resembling human plexogenic arteriopathy with intimal hyperplasia leading to occlusion of the arterial lumen. To assess the pathophysiological significance of this observation, immunohistochemistry was applied to quantitatively analyze S100A4/Mts1 expression in pulmonary arteries in surgical lung biopsies from children with pulmonary hypertension secondary to congenital heart disease. S100A4/Mts1 was not detected in pulmonary arteries with low-grade hypertensive lesions but was expressed in smooth muscle cells of lesions showing neointimal formation and with increased intensity in vessels with an occlusive neointima and plexiform lesions. Putative downstream targets of S100A4/Mts1 include Bax, which is pro-apoptotic, and the pro-angiogenic vascular endothelial growth factor (VEGF). The increase in S100A4/Mts1 expression precedes heightened expression of Bax in progressively severe neointimal lesions but in non-S100A4/Mts1-expressing cells. VEGF immunoreactivity did not correlate with severity of disease. The relationship of increased S100A4/Mts1 to pathologically similar lesions in the transgenic mice and patients occurs despite differences in localization (endothelial versus smooth muscle cells).


Subject(s)
Proto-Oncogene Proteins c-bcl-2 , Pulmonary Artery/pathology , S100 Proteins/biosynthesis , Adolescent , Animals , Child , Child, Preschool , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression , Heart Defects, Congenital/complications , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Immunohistochemistry , Infant , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein
16.
Biochem Biophys Res Commun ; 292(4): 980-6, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11944911

ABSTRACT

The examination of insulin exocytosis at the single cell level by conventional electrophysiologic and amperometric methods possesses inherent limitations, and may not accurately reflect the morphologic events of exocytosis of the insulin granule. To overcome some of these limitations, we show by epifluorescent microscopy of a fluorescent dye, FM1-43, its incorporation into the plasma membrane and oncoming insulin granules undergoing exocytosis, and their core proteins. Using this method, we tracked exocytosis in real-time in insulinoma INS-1 and single rat islet beta cells in response to KCl and glucose. We observed both single transient and multi-stepwise increases in membrane FM1-43 fluorescence, suggesting single granule exocytosis as well as sequential and compound exocytosis, respectively. Confocal microscopy of nonpermeabilized cells shows that some of the exocytosed insulin granules labeled by the FM1-43 dye could also be labeled with insulin antibodies, suggesting prolonged openings of the fusion pores and slow dissolution of the granule core proteins on the membrane surface.


Subject(s)
Exocytosis/physiology , Islets of Langerhans/metabolism , Animals , Cell Membrane/drug effects , Cells, Cultured , Exocytosis/drug effects , Fluorescent Dyes , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulinoma/drug therapy , Insulinoma/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Male , Microscopy, Confocal , Microscopy, Fluorescence , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Secretory Vesicles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...