Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biol Chem ; 293(18): 6893-6904, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29549124

RESUMO

The voltage-dependent K+ (Kv) channel Kv2.1 is a major delayed rectifier in many secretory cells, including pancreatic ß cells. In addition, Kv2.1 has a direct role in exocytosis at an undefined step, involving SNARE proteins, that is independent of its ion-conducting pore function. Here, we elucidated the precise step in exocytosis. We previously reported that syntaxin-3 (Syn-3) is the key syntaxin that mediates exocytosis of newcomer secretory granules that spend minimal residence time on the plasma membrane before fusion. Using high-resolution total internal reflection fluorescence microscopy, we now show that Kv2.1 forms reservoir clusters on the ß-cell plasma membrane and binds Syn-3 via its C-terminal C1b domain, which recruits newcomer insulin secretory granules into this large reservoir. Upon glucose stimulation, secretory granules were released from this reservoir to replenish the pool of newcomer secretory granules for subsequent fusion, occurring just adjacent to the plasma membrane Kv2.1 clusters. C1b deletion blocked the aforementioned Kv2.1-Syn-3-mediated events and reduced fusion of newcomer secretory granules. These insights have therapeutic implications, as Kv2.1 overexpression in type-2 diabetes rat islets restored biphasic insulin secretion.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Vesículas Secretórias/metabolismo , Canais de Potássio Shab/metabolismo , Animais , Membrana Celular/metabolismo , Exocitose/fisiologia , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Microscopia de Fluorescência , Ligação Proteica , Domínios Proteicos , Proteínas Qa-SNARE/química , Ratos , Ratos Wistar , Proteínas SNARE/metabolismo , Canais de Potássio Shab/fisiologia
2.
J Biol Chem ; 292(6): 2203-2216, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28031464

RESUMO

In type-2 diabetes (T2D), severely reduced islet syntaxin-1A (Syn-1A) levels contribute to insulin secretory deficiency. We generated ß-cell-specific Syn-1A-KO (Syn-1A-ßKO) mice to mimic ß-cell Syn-1A deficiency in T2D. Glucose tolerance tests showed that Syn-1A-ßKO mice exhibited blood glucose elevation corresponding to reduced blood insulin levels. Perifusion of Syn-1A-ßKO islets showed impaired first- and second-phase glucose-stimulated insulin secretion (GSIS) resulting from reduction in readily releasable pool and granule pool refilling. To unequivocally determine the ß-cell exocytotic defects caused by Syn-1A deletion, EM and total internal reflection fluorescence microscopy showed that Syn-1A-KO ß-cells had a severe reduction in the number of secretory granules (SGs) docked onto the plasma membrane (PM) at rest and reduced SG recruitment to the PM after glucose stimulation, the latter indicating defects in replenishment of releasable pools required to sustain second-phase GSIS. Whereas reduced predocked SG fusion accounted for reduced first-phase GSIS, selective reduction of exocytosis of short-dock (but not no-dock) newcomer SGs accounted for the reduced second-phase GSIS. These Syn-1A actions on newcomer SGs were partly mediated by Syn-1A interactions with newcomer SG VAMP8.


Assuntos
Exocitose , Insulina/metabolismo , Vesículas Secretórias/metabolismo , Sintaxina 1/fisiologia , Animais , Glucose/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Sintaxina 1/genética
3.
Diabetologia ; 58(6): 1250-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25762204

RESUMO

AIMS/HYPOTHESIS: Of the four exocytotic syntaxins (Syns), much is now known about the role of Syn-1A (pre-docked secretory granules [SGs]) and Syn-3 (newcomer SGs) in insulin exocytosis. Some work was reported on Syn-4's role in biphasic glucose-stimulated insulin secretion (GSIS), but its precise role in insulin SG exocytosis remains unclear. In this paper we examine this role in human beta cells. METHODS: Endogenous function of Syn-4 in human islets was assessed by knocking down its expression with lentiviral single hairpin RNA (lenti-shRNA)-RFP. Biphasic GSIS was determined by islet perifusion assay. Single-cell analysis of exocytosis of red fluorescent protein (RFP)-positive beta cells (exhibiting near-total depletion of Syn-4) was by patch clamp capacitance measurements (Cm) and total internal reflection fluorescence microscopy (TIRFM), the latter to further assess single SG behaviour. Co-immunoprecipitations were conducted on INS-1 cells to assess exocytotic complexes. RESULTS: Syn-4 knockdown (KD) of 77% in human islets caused a concomitant reduction in cognate Munc18c expression (46%) without affecting expression of other exocytotic proteins; this resulted in reduction of GSIS in the first phase (by 42%) and the second phase (by 40%). Cm of RFP-tagged Syn-4-KD beta cells showed severe inhibition in the readily releasable pool (by 71%) and mobilisation from reserve pools (by 63%). TIRFM showed that Syn-4-KD-induced inhibition of first-phase GSIS was attributed to reduction in exocytosis of both pre-docked and newcomer SGs (which undergo minimal residence or docking time at the plasma membrane before fusion). Second-phase inhibition was attributed to reduction in newcomer SGs. Stx-4 co-immunoprecipitated Munc18c, VAMP2 and VAMP8, suggesting that these exocytotic complexes may be involved in exocytosis of pre-docked and newcomer SGs. CONCLUSIONS/INTERPRETATION: Syn-4 is involved in distinct molecular machineries that influence exocytosis of both pre-docked and newcomer SGs in a manner functionally redundant to Syn-1A and Syn-3, respectively; this underlies Syn-4's role in mediating portions of first-phase and second-phase GSIS.


Assuntos
Insulinas Bifásicas/sangue , Exocitose , Células Secretoras de Insulina/citologia , Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Humanos , Secreção de Insulina , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteínas Munc18/metabolismo , Técnicas de Patch-Clamp , Proteínas R-SNARE/metabolismo , RNA Interferente Pequeno/metabolismo , Análise de Célula Única , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Proteína Vermelha Fluorescente
4.
J Biol Chem ; 289(9): 6028-40, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24429282

RESUMO

In ß-cells, syntaxin (Syn)-1A interacts with SUR1 to inhibit ATP-sensitive potassium channels (KATP channels). PIP2 binds the Kir6.2 subunit to open KATP channels. PIP2 also modifies Syn-1A clustering in plasma membrane (PM) that may alter Syn-1A actions on PM proteins like SUR1. Here, we assessed whether the actions of PIP2 on activating KATP channels is contributed by sequestering Syn-1A from binding SUR1. In vitro binding showed that PIP2 dose-dependently disrupted Syn-1A·SUR1 complexes, corroborated by an in vivo Forster resonance energy transfer assay showing disruption of SUR1(-EGFP)/Syn-1A(-mCherry) interaction along with increased Syn-1A cluster formation. Electrophysiological studies of rat ß-cells, INS-1, and SUR1/Kir6.2-expressing HEK293 cells showed that PIP2 dose-dependent activation of KATP currents was uniformly reduced by Syn-1A. To unequivocally distinguish between PIP2 actions on Syn-1A and Kir6.2, we employed several strategies. First, we showed that PIP2-insensitive Syn-1A-5RK/A mutant complex with SUR1 could not be disrupted by PIP2, consequently reducing PIP2 activation of KATP channels. Next, Syn-1A·SUR1 complex modulation of KATP channels could be observed at a physiologically low PIP2 concentration that did not disrupt the Syn-1A·SUR1 complex, compared with higher PIP2 concentrations acting directly on Kir6.2. These effects were specific to PIP2 and not observed with physiologic concentrations of other phospholipids. Finally, depleting endogenous PIP2 with polyphosphoinositide phosphatase synaptojanin-1, known to disperse Syn-1A clusters, freed Syn-1A from Syn-1A clusters to bind SUR1, causing inhibition of KATP channels that could no longer be further inhibited by exogenous Syn-1A. These results taken together indicate that PIP2 affects islet ß-cell KATP channels not only by its actions on Kir6.2 but also by sequestering Syn-1A to modulate Syn-1A availability and its interactions with SUR1 on PM.


Assuntos
Células Secretoras de Insulina/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Sulfonilureias/metabolismo , Sintaxina 1/metabolismo , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Células Secretoras de Insulina/citologia , Masculino , Mutação , Fosfatidilinositol 4,5-Difosfato/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Ratos , Ratos Wistar , Receptores de Sulfonilureias/genética , Sintaxina 1/genética
5.
Cell Rep Med ; 4(5): 101051, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37196633

RESUMO

Alterations in the microbiome correlate with improved metabolism in patients following bariatric surgery. While fecal microbiota transplantation (FMT) from obese patients into germ-free (GF) mice has suggested a significant role of the gut microbiome in metabolic improvements following bariatric surgery, causality remains to be confirmed. Here, we perform paired FMT from the same obese patients (BMI > 40; four patients), pre- and 1 or 6 months post-Roux-en-Y gastric bypass (RYGB) surgery, into Western diet-fed GF mice. Mice colonized by FMT from patients' post-surgery stool exhibit significant changes in microbiota composition and metabolomic profiles and, most importantly, improved insulin sensitivity compared with pre-RYGB FMT mice. Mechanistically, mice harboring the post-RYGB microbiome show increased brown fat mass and activity and exhibit increased energy expenditure. Moreover, improvements in immune homeostasis within the white adipose tissue are also observed. Altogether, these findings point to a direct role for the gut microbiome in mediating improved metabolic health post-RYGB surgery.


Assuntos
Cirurgia Bariátrica , Microbioma Gastrointestinal , Resistência à Insulina , Camundongos , Animais , Tecido Adiposo Marrom , Obesidade/cirurgia , Metabolismo Energético
6.
Nat Commun ; 13(1): 6512, 2022 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-36316316

RESUMO

Enhancing pancreatic ß-cell secretion is a primary therapeutic target for type-2 diabetes (T2D). Syntaxin-2 (Stx2) has just been identified to be an inhibitory SNARE for insulin granule exocytosis, holding potential as a treatment for T2D, yet its molecular underpinnings remain unclear. We show that excessive Stx2 recruitment to raft-like granule docking sites at higher binding affinity than pro-fusion syntaxin-1A effectively competes for and inhibits fusogenic SNARE machineries. Depletion of Stx2 in human ß-cells improves insulin secretion by enhancing trans-SNARE complex assembly and cis-SNARE disassembly. Using a genetically-encoded reporter, glucose stimulation is shown to induce Stx2 flipping across the plasma membrane, which relieves its suppression of cytoplasmic fusogenic SNARE complexes to promote insulin secretion. Targeting the flipping efficiency of Stx2 profoundly modulates secretion, which could restore the impaired insulin secretion in diabetes. Here, we show that Stx2 acts to assist this precise tuning of insulin secretion in ß-cells, including in diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Insulina , Humanos , Sintaxina 1/genética , Sintaxina 1/metabolismo , Insulina/metabolismo , Exocitose/fisiologia , Proteínas SNARE/metabolismo , Membrana Celular/metabolismo
7.
Autophagy ; 17(10): 3068-3081, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33213278

RESUMO

Intrapancreatic trypsin activation by dysregulated macroautophagy/autophagy and pathological exocytosis of zymogen granules (ZGs), along with activation of inhibitor of NFKB/NF-κB kinase (IKK) are necessary early cellular events in pancreatitis. How these three pancreatitis events are linked is unclear. We investigated how SNAP23 orchestrates these events leading to pancreatic acinar injury. SNAP23 depletion was by knockdown (SNAP23-KD) effected by adenovirus-shRNA (Ad-SNAP23-shRNA/mCherry) treatment of rodent and human pancreatic slices and in vivo by infusion into rat pancreatic duct. In vitro pancreatitis induction by supraphysiological cholecystokinin (CCK) or ethanol plus low-dose CCK were used to assess SNAP23-KD effects on exocytosis and autophagy. Pancreatitis stimuli resulted in SNAP23 translocation from its native location at the plasma membrane to autophagosomes, where SNAP23 would bind and regulate STX17 (syntaxin17) SNARE complex-mediated autophagosome-lysosome fusion. This SNAP23 relocation was attributed to IKBKB/IKKß-mediated SNAP23 phosphorylation at Ser95 Ser120 in rat and Ser120 in human, which was blocked by IKBKB/IKKß inhibitors, and confirmed by the inability of IKBKB/IKKß phosphorylation-disabled SNAP23 mutant (Ser95A Ser120A) to bind STX17 SNARE complex. SNAP23-KD impaired the assembly of STX4-driven basolateral exocytotic SNARE complex and STX17-driven SNARE complex, causing respective reduction of basolateral exocytosis of ZGs and autolysosome formation, with consequent reduction in trypsinogen activation in both compartments. Consequently, pancreatic SNAP23-KD rats were protected from caerulein and alcoholic pancreatitis. This study revealed the roles of SNAP23 in mediating pathological basolateral exocytosis and IKBKB/IKKß's involvement in autolysosome formation, both where trypsinogen activation would occur to cause pancreatitis. SNAP23 is a strong candidate to target for pancreatitis therapy.Abbreviations: AL: autolysosome; AP: acute pancreatitis; AV: autophagic vacuole; CCK: cholecystokinin; IKBKB/IKKß: inhibitor of nuclear factor kappa B kinase subunit beta; SNAP23: synaptosome associated protein 23; SNARE: soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein receptor; STX: syntaxin; TAP: trypsinogen activation peptide; VAMP: vesicle associated membrane protein; ZG: zymogen granule.


Assuntos
Pancreatite , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Doença Aguda , Animais , Autofagia , Exocitose , Humanos , Lisossomos , Pâncreas , Pancreatite/genética , Pancreatite/prevenção & controle , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Ratos , Tripsina/farmacologia , Proteínas de Transporte Vesicular
8.
JCI Insight ; 5(3)2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32051343

RESUMO

SNAP23 is the ubiquitous SNAP25 isoform that mediates secretion in non-neuronal cells, similar to SNAP25 in neurons. However, some secretory cells like pancreatic islet ß cells contain an abundance of both SNAP25 and SNAP23, where SNAP23 is believed to play a redundant role to SNAP25. We show that SNAP23, when depleted in mouse ß cells in vivo and human ß cells (normal and type 2 diabetes [T2D] patients) in vitro, paradoxically increased biphasic glucose-stimulated insulin secretion corresponding to increased exocytosis of predocked and newcomer insulin granules. Such effects on T2D Goto-Kakizaki rats improved glucose homeostasis that was superior to conventional treatment with sulfonylurea glybenclamide. SNAP23, although fusion competent in slower secretory cells, in the context of ß cells acts as a weak partial fusion agonist or inhibitory SNARE. Here, SNAP23 depletion promotes SNAP25 to bind calcium channels more quickly and longer where granule fusion occurs to increase exocytosis efficiency. ß Cell SNAP23 antagonism is a strategy to treat diabetes.


Assuntos
Canais de Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exocitose , Insulina/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Animais , Glucose/metabolismo , Homeostase , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Ratos
9.
EBioMedicine ; 16: 262-274, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28163042

RESUMO

Reduced pancreatic islet levels of Munc18a/SNARE complex proteins have been postulated to contribute to the deficient glucose-stimulated insulin secretion (GSIS) in type-2 diabetes (T2D). Whereas much previous work has purported Munc18a/SNARE complex (Syntaxin-1A/VAMP-2/SNAP25) to be primarily involved in predocked secretory granule (SG) fusion, less is known about newcomer SGs that undergo minimal docking time at the plasma membrane before fusion. Newcomer SG fusion has been postulated to involve a distinct SM/SNARE complex (Munc18b/Syntaxin-3/VAMP8/SNAP25), whose levels we find also reduced in islets of T2D humans and T2D Goto-Kakizaki (GK) rats. Munc18b overexpression by adenovirus infection (Ad-Munc18b), by increasing assembly of Munc18b/SNARE complexes, mediated increased fusion of not only newcomer SGs but also predocked SGs in T2D human and GK rat islets, resulting in rescue of the deficient biphasic GSIS. Infusion of Ad-Munc18b into GK rat pancreas led to sustained improvement in glucose homeostasis. However, Munc18b overexpression in normal islets increased only newcomer SG fusion. Therefore, Munc18b could potentially be deployed in human T2D to rescue the deficient GSIS.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Munc18/metabolismo , Idoso , Animais , Western Blotting , Diabetes Mellitus Tipo 2/genética , Feminino , Humanos , Secreção de Insulina , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Complexos Multiproteicos/metabolismo , Proteínas Munc18/genética , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Ratos Endogâmicos , Proteína 25 Associada a Sinaptossoma/metabolismo
10.
Diabetes ; 66(4): 948-959, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28115395

RESUMO

Of the four syntaxins specialized for exocytosis, syntaxin (Syn)-2 is the least understood. In this study, we used Syn-2/epimorphin knockout mice to examine the role of Syn-2 in insulin secretory granule (SG) exocytosis. Unexpectedly, Syn-2 knockout mice exhibited paradoxical superior glucose homeostasis resulting from an enhanced insulin secretion. This was confirmed in vitro by pancreatic islet perifusion showing an amplified biphasic glucose-stimulated insulin secretion arising from an increase in size of the readily releasable pool of insulin SGs and enhanced SG pool refilling. The increase in insulin exocytosis was attributed mainly to an enhanced recruitment of the larger pool of newcomer SGs that undergoes no residence time on plasma membrane before fusion and, to a lesser extent, also the predocked SGs. Consistently, Syn-2 depletion resulted in a stimulation-induced increase in abundance of exocytotic complexes we previously demonstrated as mediating the fusion of newcomer SGs (Syn-3/VAMP8/SNAP25/Munc18b) and predocked SGs (Syn-1A/VAMP2/SNAP25/Muncn18a). This work is the first to show in mammals that Syn-2 could function as an inhibitory SNARE protein that, when relieved, could promote exocytosis in pancreatic islet ß-cells. Thus, Syn-2 may serve as a potential target to treat diabetes.


Assuntos
Exocitose/genética , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas SNARE/metabolismo , Sintaxina 1/genética , Animais , Western Blotting , Imunoprecipitação , Secreção de Insulina , Camundongos , Camundongos Knockout , Proteínas Munc18/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Vesículas Secretórias/metabolismo , Frações Subcelulares , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo
11.
PLoS One ; 11(2): e0147862, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26848587

RESUMO

Syntaxin (Syn)-1A mediates exocytosis of predocked insulin-containing secretory granules (SGs) during first-phase glucose-stimulated insulin secretion (GSIS) in part via its interaction with plasma membrane (PM)-bound L-type voltage-gated calcium channels (Cav). In contrast, Syn-3 mediates exocytosis of newcomer SGs that accounts for second-phase GSIS. We now hypothesize that the newcomer SG Syn-3 preferentially binds and modulates R-type Cav opening, which was postulated to mediate second-phase GSIS. Indeed, glucose-stimulation of pancreatic islet ß-cell line INS-1 induced a predominant increase in interaction between Syn-3 and Cavα1 pore-forming subunits of R-type Cav2.3 and to lesser extent L-type Cavs, while confirming the preferential interactions between Syn-1A with L-type (Cav1.2, Cav1.3) Cavs. Consistently, direct binding studies employing heterologous HEK cells confirmed that Syn-3 preferentially binds Cav2.3, whereas Syn-1A prefers L-type Cavs. We then used siRNA knockdown (KD) of Syn-3 in INS-1 to study the endogenous modulatory actions of Syn-3 on Cav channels. Syn-3 KD enhanced Ca2+ currents by 46% attributed mostly to R- and L-type Cavs. Interestingly, while the transmembrane domain of Syn-1A is the putative functional domain modulating Cav activity, it is the cytoplasmic domain of Syn-3 that appears to modulate Cav activity. We conclude that Syn-3 may mimic Syn-1A in the ability to bind and modulate Cavs, but preferring Cav2.3 to perhaps participate in triggering fusion of newcomer insulin SGs during second-phase GSIS.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo R/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Linhagem Celular , Humanos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/genética , Sintaxina 1/química , Sintaxina 1/metabolismo
12.
Diabetes ; 65(7): 1962-76, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207520

RESUMO

Synaptotagmin (Syt)-7, a major component of the exocytotic machinery in neurons, is also the major Syt in rodent pancreatic ß-cells shown to mediate glucose-stimulated insulin secretion (GSIS). However, Syt-7's precise exocytotic actions in ß-cells remain unknown. We show that Syt-7 is abundant in human ß-cells. Adenovirus-short hairpin RNA knockdown (KD) of Syt-7 in human islets reduced first- and second-phase GSIS attributed to the reduction of exocytosis of predocked and newcomer insulin secretory granules (SGs). Glucose stimulation expectedly induced Syt-7 association in a Ca(2+)-dependent manner with syntaxin-3 and syntaxin-1A soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes known to mediate exocytosis of newcomer and predocked SGs, respectively. However, Syt-7-KD did not disrupt SNARE complex assembly. Instead, electron microscopy analysis showed that Syt-7-KD reduced the recruitment of SGs to the plasma membrane after glucose-stimulated depletion, which could not be rescued by glucagon-like peptide 1 pretreatment. To assess the possibility that this new action of Syt-7 on SG recruitment may involve calmodulin (CaM), pretreatment of islets with CaM blocker calmidazolium showed effects very similar to those of Syt-7-KD. Syt-7 therefore plays a novel more dominant function in the replenishment of releasable SG pools in human ß-cells than its previously purported role in exocytotic fusion per se.


Assuntos
Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Vesículas Secretórias/metabolismo , Sinaptotagminas/metabolismo , Adulto , Idoso , Animais , Cálcio/metabolismo , Exocitose/efeitos dos fármacos , Feminino , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucose/farmacologia , Humanos , Imidazóis/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Proteínas Qa-SNARE/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas SNARE/metabolismo
13.
Pancreas ; 43(2): 240-4, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24518502

RESUMO

OBJECTIVE: Pancreatic islets are notoriously difficult to efficiently transduce genes with viruses whether in vivo or ex vivo, the latter only transducing superficial layers of the islet. To improve efficiency of transduction, we explored surgical approaches to virus delivery in vivo. METHODS: A technique was developed for retrograde surgical perfusion into the rat biliopancreatic duct with a test adenovirus containing a construct coexpressing green fluorescent protein, the latter for detection of infected cells. RESULTS: Pancreatic islets isolated after acute pancreatic infusion and cultured for 2 days showed expression in the entire islet and in almost all islets. When rats were recovered from the surgery, and then islets isolated at 1 and 8 weeks after surgery, we continued to see extensive islet green fluorescent protein expression, albeit at more reduced levels at 8 weeks. CONCLUSIONS: This strategy of surgical pancreatic ductal perfusion of viruses is an effective way to transduce or reduce gene expression in pancreatic islets for both acute and chronic study.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução Genética/métodos , Adenoviridae/genética , Animais , Sistema Biliar , Sistemas de Liberação de Medicamentos/métodos , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Bombas de Infusão , Masculino , Microscopia Confocal , Ductos Pancreáticos , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Reprodutibilidade dos Testes
14.
Diabetes ; 62(8): 2968-77, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23630299

RESUMO

Impaired counterregulation during hypoglycemia in type 1 diabetes (T1D) is partly attributable to inadequate glucagon secretion. Intra-islet somatostatin (SST) suppression of hypoglycemia-stimulated α-cell glucagon release plays an important role. We hypothesized that hypoglycemia can be prevented in autoimmune T1D by SST receptor type 2 (SSTR2) antagonism of α-cells, which relieve SSTR2 inhibition, thereby increasing glucagon secretion. Diabetic biobreeding diabetes-prone (BBDP) rats mimic insulin-dependent human autoimmune T1D, whereas nondiabetic BBDP rats mimic prediabetes. Diabetic and nondiabetic rats underwent a 3-h infusion of vehicle compared with SSTR2 antagonist (SSTR2a) during insulin-induced hypoglycemia clamped at 3 ± 0.5 mmol/L. Diabetic rats treated with SSTR2a needed little or no glucose infusion compared with untreated rats. We attribute this effect to SSTR2a restoration of the attenuated glucagon response. Direct effects of SSTR2a on α-cells was assessed by resecting the pancreas, which was cut into fine slices and subjected to perifusion to monitor glucagon release. SSTR2a treatment enhanced low-glucose-stimulated glucagon and corticosterone secretion to normal levels in diabetic rats. SSTR2a had similar effects in vivo in nondiabetic rats and promoted glucagon secretion from nondiabetic rat and human pancreas slices. We conclude that SST contributes to impaired glucagon responsiveness to hypoglycemia in autoimmune T1D. SSTR2a treatment can fully restore hypoglycemia-stimulated glucagon release sufficient to attain normoglycemia in both diabetic and prediabetic stages.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Glucagon/metabolismo , Pâncreas/efeitos dos fármacos , Estado Pré-Diabético/metabolismo , Receptores de Somatostatina/antagonistas & inibidores , Animais , Catecolaminas/metabolismo , Corticosterona/metabolismo , Células Secretoras de Glucagon/metabolismo , Técnica Clamp de Glucose , Humanos , Insulina/metabolismo , Secreção de Insulina , Masculino , Pâncreas/metabolismo , Peptídeos Cíclicos/farmacologia , Ratos , Somatostatina/metabolismo
15.
Diabetes ; 62(7): 2416-28, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23423569

RESUMO

Sec1/Munc18 proteins facilitate the formation of trans-SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes that mediate fusion of secretory granule (SG) with plasma membrane (PM). The capacity of pancreatic ß-cells to exocytose insulin becomes compromised in diabetes. ß-Cells express three Munc18 isoforms of which the role of Munc18b is unknown. We found that Munc18b depletion in rat islets disabled SNARE complex formation formed by syntaxin (Syn)-2 and Syn-3. Two-photon imaging analysis revealed in Munc18b-depleted ß-cells a 40% reduction in primary exocytosis (SG-PM fusion) and abrogation of almost all sequential SG-SG fusion, together accounting for a 50% reduction in glucose-stimulated insulin secretion (GSIS). In contrast, gain-of-function expression of Munc18b wild-type and, more so, dominant-positive K314L/R315L mutant promoted the assembly of cognate SNARE complexes, which caused potentiation of biphasic GSIS. We found that this was attributed to a more than threefold enhancement of both primary exocytosis and sequential SG-SG fusion, including long-chain fusion (6-8 SGs) not normally (2-3 SG fusion) observed. Thus, Munc18b-mediated exocytosis may be deployed to increase secretory efficiency of SGs in deeper cytosolic layers of ß-cells as well as additional primary exocytosis, which may open new avenues of therapy development for diabetes.


Assuntos
Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Munc18/metabolismo , Vesículas Secretórias/metabolismo , Animais , Secreção de Insulina , Masculino , Proteínas Munc18/genética , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Ratos , Ratos Sprague-Dawley , Sintaxina 1/genética , Sintaxina 1/metabolismo
16.
Cell Metab ; 16(2): 238-49, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22841572

RESUMO

Optimal insulin secretion required to maintain glucose homeostasis is the summation of total pancreatic islet ß cell mass and intrinsic secretory capacity of individual ß cells, which are regulated by distinct mechanisms that could be amplified by glucagon-like-peptide-1 (GLP-1). Because of these actions of GLP-1 on islet ß cells, GLP-1 has been deployed to treat diabetes. We employed SNARE protein VAMP8-null mice to demonstrate that VAMP8 mediates insulin granule recruitment to the plasma membrane, which partly accounts for GLP-1 potentiation of glucose-stimulated insulin secretion. VAMP8-null mice also exhibited increased islet ß cell mass from increased ß cell mitosis, with ß cell proliferative activity greatly amplified by GLP-1. Thus, despite the ß cell exocytotic defect, VAMP8-null mice have an increased total insulin secretory capacity, which improved glucose homeostasis. We conclude that these VAMP8-mediated events partly underlie the therapeutic actions of GLP-1 on insulin secretion and ß cell growth.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Exocitose/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Proteínas R-SNARE/metabolismo , Análise de Variância , Animais , Western Blotting , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Imuno-Histoquímica , Imunoprecipitação , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Proteínas R-SNARE/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA