Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Diabetologia ; 67(8): 1642-1662, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38743124

RESUMO

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).


Assuntos
Haploinsuficiência , Células Secretoras de Insulina , Fatores de Transcrição de Fator Regulador X , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição de Fator Regulador X/genética , Fatores de Transcrição de Fator Regulador X/metabolismo , Animais , Humanos , Camundongos , Diferenciação Celular/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Predisposição Genética para Doença , Feminino , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Tecido Nervoso
2.
Proc Natl Acad Sci U S A ; 117(5): 2484-2495, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964806

RESUMO

Circadian clocks operative in pancreatic islets participate in the regulation of insulin secretion in humans and, if compromised, in the development of type 2 diabetes (T2D) in rodents. Here we demonstrate that human islet α- and ß-cells that bear attenuated clocks exhibit strongly disrupted insulin and glucagon granule docking and exocytosis. To examine whether compromised clocks play a role in the pathogenesis of T2D in humans, we quantified parameters of molecular clocks operative in human T2D islets at population, single islet, and single islet cell levels. Strikingly, our experiments reveal that islets from T2D patients contain clocks with diminished circadian amplitudes and reduced in vitro synchronization capacity compared to their nondiabetic counterparts. Moreover, our data suggest that islet clocks orchestrate temporal profiles of insulin and glucagon secretion in a physiological context. This regulation was disrupted in T2D subjects, implying a role for the islet cell-autonomous clocks in T2D progression. Finally, Nobiletin, an agonist of the core-clock proteins RORα/γ, boosted both circadian amplitude of T2D islet clocks and insulin secretion by these islets. Our study emphasizes a link between the circadian clockwork and T2D and proposes that clock modulators hold promise as putative therapeutic agents for this frequent disorder.


Assuntos
Ritmo Circadiano/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 2/fisiopatologia , Exocitose/efeitos dos fármacos , Feminino , Flavonas/farmacologia , Humanos , Técnicas In Vitro , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
3.
Semin Cell Dev Biol ; 103: 20-30, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32085965

RESUMO

The cytoplasmic Ca2+ concentration ([Ca2+]cyt) regulates a vast number of cellular functions, including insulin secretion from beta cells. The major physiological insulin secretagogue, glucose, triggers [Ca2+]cyt oscillations in beta cells. Synchronization of the oscillations among the beta cells within an islet underlies the generation of pulsatile insulin secretion. This review describes the mechanisms generating [Ca2+]cyt oscillations, the interactions between [Ca2+]cyt and cell metabolism, as well as the contribution of various organelles to the shaping of [Ca2+]cyt signals and insulin secretion. It also discusses how Ca2+ signals are coordinated and spread throughout the islets and data indicating that altered Ca2+ signaling is associated with beta cell dysfunction and development of type 2 diabetes.


Assuntos
Sinalização do Cálcio/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Humanos
4.
J Biol Chem ; 295(16): 5419-5426, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32156704

RESUMO

Hypersecretion of glucagon from pancreatic α-cells strongly contributes to diabetic hyperglycemia. Moreover, failure of α-cells to increase glucagon secretion in response to falling blood glucose concentrations compromises the defense against hypoglycemia, a common complication in diabetes therapy. However, the mechanisms underlying glucose regulation of glucagon secretion are poorly understood and likely involve both α-cell-intrinsic and intraislet paracrine signaling. Among paracrine factors, glucose-stimulated release of the GABA metabolite γ-hydroxybutyric acid (GHB) from pancreatic ß-cells might mediate glucose suppression of glucagon release via GHB receptors on α-cells. However, the direct effects of GHB on α-cell signaling and glucagon release have not been investigated. Here, we found that GHB (4-10 µm) lacked effects on the cytoplasmic concentrations of the secretion-regulating messengers Ca2+ and cAMP in mouse α-cells. Glucagon secretion from perifused mouse islets was also unaffected by GHB at both 1 and 7 mm glucose. The GHB receptor agonist 3-chloropropanoic acid and the antagonist NCS-382 had no effects on glucagon secretion and did not affect stimulation of secretion induced by a drop in glucose from 7 to 1 mm Inhibition of endogenous GHB formation with the GABA transaminase inhibitor vigabatrin also failed to influence glucagon secretion at 1 mm glucose and did not prevent the suppressive effect of 7 mm glucose. In human islets, GHB tended to stimulate glucagon secretion at 1 mm glucose, an effect mimicked by 3-chloropropanoic acid. We conclude that GHB does not mediate the inhibitory effect of glucose on glucagon secretion.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Oxibato de Sódio/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Benzocicloeptenos/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Feminino , GABAérgicos/farmacologia , Células Secretoras de Glucagon/efeitos dos fármacos , Glucose/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Propionatos/farmacologia , Vigabatrina/farmacologia
5.
FASEB J ; 33(1): 88-100, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29957057

RESUMO

Zinc finger BED domain containing protein 6 ( Zbed6) has evolved from a domesticated DNA transposon and encodes a transcription factor unique to placental mammals. The aim of the present study was to investigate further the role of ZBED6 in insulin-producing cells, using mouse MIN6 cells, and to evaluate the effects of Zbed6 knockdown on basal ß-cell functions, such as morphology, transcriptional regulation, insulin content, and release. Zbed6-silenced cells and controls were characterized with a range of methods, including RNA sequencing, chromatin immunoprecipitation sequencing, insulin content and release, subplasma membrane Ca2+ measurements, cAMP determination, and morphologic studies. More than 700 genes showed differential expression in response to Zbed6 knockdown, which was paralleled by increased capacity to generate cAMP, as well as by augmented subplasmalemmal calcium concentration and insulin secretion in response to glucose stimulation. We identified >4000 putative ZBED6-binding sites in the MIN6 genome, with an enrichment of ZBED6 sites at upregulated genes, such as the ß-cell transcription factors v-maf musculoaponeurotic fibrosarcoma oncogene homolog A and Nk6 homeobox 1. We also observed altered morphology/growth patterns, as indicated by increased cell clustering, and in the appearance of axon-like Neurofilament, medium polypeptide and tubulin ß 3, class III-positive protrusions. We conclude that ZBED6 acts as a transcriptional regulator in MIN6 cells and that its activity suppresses insulin production, cell aggregation, and neuronal-like differentiation.-Wang, X., Jiang, L., Wallerman, O., Younis, S., Yu, Q., Klaesson, A., Tengholm, A., Welsh, N., Andersson, L. ZBED6 negatively regulates insulin production, neuronal differentiation, and cell aggregation in MIN6 cells.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/patologia , Insulina/metabolismo , Insulinoma/patologia , Neurônios/patologia , Neoplasias Pancreáticas/patologia , Proteínas Repressoras/metabolismo , Animais , Sítios de Ligação , Adesão Celular , Agregação Celular , Regulação da Expressão Gênica , Inativação Gênica , Glucose/administração & dosagem , Sequenciamento de Nucleotídeos em Larga Escala , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Camundongos , Neurônios/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transcrição Gênica , Células Tumorais Cultivadas
6.
Diabetologia ; 62(7): 1212-1224, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30953108

RESUMO

AIMS/HYPOTHESIS: Glucagon is critical for normal glucose homeostasis and aberrant secretion of the hormone aggravates dysregulated glucose control in diabetes. However, the mechanisms by which glucose controls glucagon secretion from pancreatic alpha cells remain elusive. The aim of this study was to investigate the role of the intracellular messenger cAMP in alpha-cell-intrinsic glucose regulation of glucagon release. METHODS: Subplasmalemmal cAMP and Ca2+ concentrations were recorded in isolated and islet-located alpha cells using fluorescent reporters and total internal reflection microscopy. Glucagon secretion from mouse islets was measured using ELISA. RESULTS: Glucose induced Ca2+-independent alterations of the subplasmalemmal cAMP concentration in alpha cells that correlated with changes in glucagon release. Glucose-lowering-induced stimulation of glucagon secretion thus corresponded to an elevation in cAMP that was independent of paracrine signalling from insulin or somatostatin. Imposed cAMP elevations stimulated glucagon secretion and abolished inhibition by glucose elevation, while protein kinase A inhibition mimicked glucose suppression of glucagon release. CONCLUSIONS/INTERPRETATION: Glucose concentrations in the hypoglycaemic range control glucagon secretion by directly modulating the cAMP concentration in alpha cells independently of paracrine influences. These findings define a novel mechanism for glucose regulation of glucagon release that underlies recovery from hypoglycaemia and may be disturbed in diabetes.


Assuntos
Cálcio/metabolismo , AMP Cíclico/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Glucose/metabolismo , Hipoglicemia/metabolismo , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Somatostatina/metabolismo
7.
J Biol Chem ; 291(29): 14986-95, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226533

RESUMO

PKC signaling has been implicated in the regulation of many cell functions, including metabolism, cell death, proliferation, and secretion. Activation of conventional and novel PKC isoforms is associated with their Ca(2+)- and/or diacylglycerol (DAG)-dependent translocation to the plasma membrane. In ß cells, exocytosis of insulin granules evokes brief (<10 s) local DAG elevations ("spiking") at the plasma membrane because of autocrine activation of P2Y1 purinoceptors by ATP co-released with insulin. Using total internal reflection microscopy, fluorescent protein-tagged PKCs, and signaling biosensors, we investigated whether DAG spiking causes membrane recruitment of PKCs and whether different classes of PKCs show characteristic responses. Glucose stimulation of MIN6 cells triggered DAG spiking with concomitant repetitive translocation of the novel isoforms PKCδ, PKCϵ, and PKCη. The conventional PKCα, PKCßI, and PKCßII isoforms showed a more complex pattern with both rapid and slow translocation. K(+) depolarization-induced PKCϵ translocation entirely mirrored DAG spiking, whereas PKCßI translocation showed a sustained component, reflecting the subplasma membrane Ca(2+) concentration ([Ca(2+)]pm), with additional effect during DAG spikes. Interference with DAG spiking by purinoceptor inhibition prevented intermittent translocation of PKCs and reduced insulin secretion but did not affect [Ca(2+)]pm elevation or sustained PKCßI translocation. The muscarinic agonist carbachol induced pronounced transient PKCßI translocation and sustained recruitment of PKCϵ. When rise of [Ca(2+)]pm was prevented, the carbachol-induced DAG and PKCϵ responses were somewhat reduced, but PKCßI translocation was completely abolished. We conclude that exocytosis-induced DAG spikes efficiently recruit both conventional and novel PKCs to the ß cell plasma membrane. PKC signaling is thus implicated in autocrine regulation of ß cell function.


Assuntos
Comunicação Autócrina , Membrana Celular/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais , Animais , Comunicação Autócrina/efeitos dos fármacos , Carbacol/farmacologia , Linhagem Celular Tumoral , Diglicerídeos/metabolismo , Glucose/farmacologia , Insulina/farmacologia , Células Secretoras de Insulina/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Isoenzimas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptores Muscarínicos/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Diabetes Obes Metab ; 19 Suppl 1: 42-53, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28466587

RESUMO

The "second messenger" archetype cAMP is one of the most important cellular signalling molecules with central functions including the regulation of insulin and glucagon secretion from the pancreatic ß- and α-cells, respectively. cAMP is generally considered as an amplifier of insulin secretion triggered by Ca2+ elevation in the ß-cells. Both messengers are also positive modulators of glucagon release from α-cells, but in this case cAMP may be the important regulator and Ca2+ have a more permissive role. The actions of cAMP are mediated by protein kinase A (PKA) and the guanine nucleotide exchange factor Epac. The present review focuses on how cAMP is regulated by nutrients, hormones and neural factors in ß- and α-cells via adenylyl cyclase-catalysed generation and phosphodiesterase-mediated degradation. We will also discuss how PKA and Epac affect ion fluxes and the secretory machinery to transduce the stimulatory effects on insulin and glucagon secretion. Finally, we will briefly describe disturbances of the cAMP system associated with diabetes and how cAMP signalling can be targeted to normalize hypo- and hypersecretion of insulin and glucagon, respectively, in diabetic patients.


Assuntos
AMP Cíclico/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sistemas do Segundo Mensageiro , Adenilil Ciclases/metabolismo , Animais , Sinalização do Cálcio , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/metabolismo , Exocitose , Células Secretoras de Glucagon/enzimologia , Glucose/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Comunicação Parácrina , Diester Fosfórico Hidrolases/metabolismo
9.
Diabetologia ; 59(9): 1928-37, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27338624

RESUMO

AIMS/HYPOTHESIS: Insufficient insulin release and hyperglucagonaemia are culprits in type 2 diabetes. Cocaine- and amphetamine-regulated transcript (CART, encoded by Cartpt) affects islet hormone secretion and beta cell survival in vitro in rats, and Cart (-/-) mice have diminished insulin secretion. We aimed to test if CART is differentially regulated in human type 2 diabetic islets and if CART affects insulin and glucagon secretion in vitro in humans and in vivo in mice. METHODS: CART expression was assessed in human type 2 diabetic and non-diabetic control pancreases and rodent models of diabetes. Insulin and glucagon secretion was examined in isolated islets and in vivo in mice. Ca(2+) oscillation patterns and exocytosis were studied in mouse islets. RESULTS: We report an important role of CART in human islet function and glucose homeostasis in mice. CART was found to be expressed in human alpha and beta cells and in a subpopulation of mouse beta cells. Notably, CART expression was several fold higher in islets of type 2 diabetic humans and rodents. CART increased insulin secretion in vivo in mice and in human and mouse islets. Furthermore, CART increased beta cell exocytosis, altered the glucose-induced Ca(2+) signalling pattern in mouse islets from fast to slow oscillations and improved synchronisation of the oscillations between different islet regions. Finally, CART reduced glucagon secretion in human and mouse islets, as well as in vivo in mice via diminished alpha cell exocytosis. CONCLUSIONS/INTERPRETATION: We conclude that CART is a regulator of glucose homeostasis and could play an important role in the pathophysiology of type 2 diabetes. Based on the ability of CART to increase insulin secretion and reduce glucagon secretion, CART-based agents could be a therapeutic modality in type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Glucagon/metabolismo , Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Western Blotting , Sinalização do Cálcio/fisiologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/sangue , Eletrofisiologia , Exocitose/genética , Exocitose/fisiologia , Feminino , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Homeostase , Humanos , Imuno-Histoquímica , Hibridização In Situ , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Reação em Cadeia da Polimerase em Tempo Real
10.
Pflugers Arch ; 468(10): 1765-77, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27539300

RESUMO

The islets of Langerhans contain different types of endocrine cells, which are crucial for glucose homeostasis. ß- and α-cells that release insulin and glucagon, respectively, are most abundant, whereas somatostatin-producing δ-cells and particularly pancreatic polypeptide-releasing PP-cells are more scarce. Studies of islet cell function are hampered by difficulties to identify the different cell types, especially in live-cell imaging experiments when immunostaining is unsuitable. The aim of the present study was to create a set of vectors for fluorescent protein expression with cell-type-specific promoters and evaluate their applicability in functional islet imaging. We constructed six adenoviral vectors for expression of red and green fluorescent proteins controlled by the insulin, preproglucagon, somatostatin, or pancreatic polypeptide promoters. After transduction of mouse and human islets or dispersed islet cells, a majority of the fluorescent cells also immunostained for the appropriate hormone. Recordings of the sub-plasma membrane Ca(2+) and cAMP concentrations with a fluorescent indicator and a protein biosensor, respectively, showed that labeled cells respond to glucose and other modulators of secretion and revealed a striking variability in Ca(2+) signaling among α-cells. The measurements allowed comparison of the phase relationship of Ca(2+) oscillations between different types of cells within intact islets. We conclude that the fluorescent protein vectors allow easy identification of specific islet cell types and can be used in live-cell imaging together with organic dyes and genetically encoded biosensors. This approach will facilitate studies of normal islet physiology and help to clarify molecular defects and disturbed cell interactions in diabetic islets.


Assuntos
Vetores Genéticos/genética , Ilhotas Pancreáticas/metabolismo , Proteínas Luminescentes/genética , Adenoviridae/genética , Animais , Sinalização do Cálcio , Feminino , Humanos , Ilhotas Pancreáticas/citologia , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Vermelha Fluorescente
11.
EMBO J ; 31(20): 3991-4004, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-22940692

RESUMO

Endocrine release of insulin principally controls glucose homeostasis. Nutrient-induced exocytosis of insulin granules from pancreatic ß-cells involves ion channels and mobilization of Ca(2+) and cyclic AMP (cAMP) signalling pathways. Whole-animal physiology, islet studies and live-ß-cell imaging approaches reveal that ablation of the kinase/phosphatase anchoring protein AKAP150 impairs insulin secretion in mice. Loss of AKAP150 impacts L-type Ca(2+) currents, and attenuates cytoplasmic accumulation of Ca(2+) and cAMP in ß-cells. Yet surprisingly AKAP150 null animals display improved glucose handling and heightened insulin sensitivity in skeletal muscle. More refined analyses of AKAP150 knock-in mice unable to anchor protein kinase A or protein phosphatase 2B uncover an unexpected observation that tethering of phosphatases to a seven-residue sequence of the anchoring protein is the predominant molecular event underlying these metabolic phenotypes. Thus anchored signalling events that facilitate insulin secretion and glucose homeostasis may be set by AKAP150 associated phosphatase activity.


Assuntos
Proteínas de Ancoragem à Quinase A/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Resistência à Insulina/genética , Proteínas de Membrana/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/deficiência , Proteínas de Ancoragem à Quinase A/genética , Motivos de Aminoácidos , Animais , Calcineurina/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , AMP Cíclico/fisiologia , Glucose/farmacologia , Homeostase/efeitos dos fármacos , Insulina/metabolismo , Insulina/farmacologia , Secreção de Insulina , Insulinoma/patologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Fígado/enzimologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Músculo Esquelético/enzimologia , Neoplasias Pancreáticas/patologia , Mapeamento de Interação de Proteínas , Proteínas Quinases/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia , Deleção de Sequência , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
12.
FASEB J ; 29(8): 3379-88, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25911612

RESUMO

Cytoplasmic ATP and Ca(2+) are implicated in current models of glucose's control of glucagon and insulin secretion from pancreatic α- and ß-cells, respectively, but little is known about ATP and its relation to Ca(2+) in α-cells. We therefore expressed the fluorescent ATP biosensor Perceval in mouse pancreatic islets and loaded them with a Ca(2+) indicator. With total internal reflection fluorescence microscopy, we recorded subplasma membrane concentrations of Ca(2+) and ATP ([Ca(2+)]pm; [ATP]pm) in superficial α- and ß-cells of intact islets and related signaling to glucagon and insulin secretion by immunoassay. Consistent with ATP's controlling glucagon and insulin secretion during hypo- and hyperglycemia, respectively, the dose-response relationship for glucose-induced [ATP]pm generation was left shifted in α-cells compared to ß-cells. Both cell types showed [Ca(2+)]pm and [ATP]pm oscillations in opposite phase, probably reflecting energy-consuming Ca(2+) transport. Although pulsatile insulin and glucagon release are in opposite phase, [Ca(2+)]pm synchronized in the same phase between α- and ß-cells. This paradox can be explained by the overriding of Ca(2+) stimulation by paracrine inhibition, because somatostatin receptor blockade potently stimulated glucagon release with little effect on Ca(2+). The data indicate that an α-cell-intrinsic mechanism controls glucagon in hypoglycemia and that paracrine factors shape pulsatile secretion in hyperglycemia.


Assuntos
Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Transdução de Sinais/fisiologia , Animais , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos C57BL
13.
Am J Physiol Gastrointest Liver Physiol ; 309(8): G625-34, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26206857

RESUMO

Neuropeptide S (NPS) receptor (NPSR1) polymorphisms are associated with enteral dysmotility and inflammatory bowel disease (IBD). This study investigated the role of NPS in conjunction with nitrergic mechanisms in the regulation of intestinal motility and mucosal permeability. In rats, small intestinal myoelectric activity and luminal pressure changes in small intestine and colon, along with duodenal permeability, were studied. In human intestine, NPS and NPSR1 were localized by immunostaining. Pre- and postprandial plasma NPS was measured by ELISA in healthy and active IBD humans. Effects and mechanisms of NPS were studied in human intestinal muscle strips. In rats, NPS 100-4,000 pmol·kg(-1)·min(-1) had effects on the small intestine and colon. Low doses of NPS increased myoelectric spiking (P < 0.05). Higher doses reduced spiking and prolonged the cycle length of the migrating myoelectric complex, reduced intraluminal pressures (P < 0.05-0.01), and increased permeability (P < 0.01) through NO-dependent mechanisms. In human intestine, NPS localized at myenteric nerve cell bodies and fibers. NPSR1 was confined to nerve cell bodies. Circulating NPS in humans was tenfold below the ∼0.3 nmol/l dissociation constant (Kd) of NPSR1, with no difference between healthy and IBD subjects. In human intestinal muscle strips precontracted by bethanechol, NPS 1-1,000 nmol/l induced NO-dependent muscle relaxation (P < 0.05) that was sensitive also to tetrodotoxin (P < 0.01). In conclusion, NPS inhibits motility and increases permeability in neurocrine fashion acting through NO in the myenteric plexus in rats and humans. Aberrant signaling and upregulation of NPSR1 could potentially exacerbate dysmotility and hyperpermeability by local mechanisms in gastrointestinal functional and inflammatory reactions.


Assuntos
Motilidade Gastrointestinal/fisiologia , Mucosa Intestinal/metabolismo , Neuropeptídeos/metabolismo , Óxido Nítrico/metabolismo , Adulto , Animais , Betanecol , Biomarcadores , Regulação da Expressão Gênica/fisiologia , Humanos , Inflamação/metabolismo , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Neuropeptídeos/sangue , Neuropeptídeos/farmacologia , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Permeabilidade , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo
14.
Diabetologia ; 57(12): 2436-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25277952

RESUMO

Insulin secretory vesicles contain high concentrations of adenine nucleotides, which are co-released with insulin during exocytosis. There is strong evidence that ATP and ADP serve as autocrine messengers in pancreatic beta cells, but the functional effects and detailed mechanisms of action are under debate. In this issue of Diabetologia, Khan and colleagues (DOI: 10.1007/s00125-014-3368-8 ) present the results of their study of autocrine purinergic signalling in isolated human beta cells. Using a combination of electrophysiological techniques, Ca(2+) imaging and measurements of insulin secretion, it is demonstrated that voltage-dependent Ca(2+) influx triggers release of ATP/ADP, which activates purinergic receptors of the Gq/11-coupled P2Y1 isoform. Activation of these receptors leads to membrane depolarisation and phospholipase C-mediated mobilisation of Ca(2+) from endoplasmic reticulum stores, which amplifies the exocytosis-triggering Ca(2+) signal. In contrast, there is little evidence for involvement of ionotropic P2X receptors in the autocrine stimulation of human beta cells. This commentary discusses these findings as well as various functional and therapeutic implications of the complex purinergic signalling network in the pancreatic islet.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Humanos
15.
J Cell Sci ; 125(Pt 21): 5084-95, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22946044

RESUMO

Specificity and versatility in cyclic AMP (cAMP) signalling are governed by the spatial localisation and temporal dynamics of the signal. Phosphodiesterases (PDEs) are important for shaping cAMP signals by hydrolyzing the nucleotide. In pancreatic ß-cells, glucose triggers sub-plasma-membrane cAMP oscillations, which are important for insulin secretion, but the mechanisms underlying the oscillations are poorly understood. Here, we investigated the role of different PDEs in the generation of cAMP oscillations by monitoring the concentration of cAMP in the sub-plasma-membrane space ([cAMP](pm)) with ratiometric evanescent wave microscopy in MIN6 cells or mouse pancreatic ß-cells expressing a fluorescent translocation biosensor. The general PDE inhibitor IBMX increased [cAMP](pm), and whereas oscillations were frequently observed at 50 µM IBMX, 300 µM-1 mM of the inhibitor caused a stable increase in [cAMP](pm). The [cAMP](pm) was nevertheless markedly suppressed by the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine, indicating IBMX-insensitive cAMP degradation. Among IBMX-sensitive PDEs, PDE3 was most important for maintaining a low basal level of [cAMP](pm) in unstimulated cells. After glucose induction of [cAMP](pm) oscillations, inhibitors of PDE1, PDE3 and PDE4 inhibitors the average cAMP level, often without disturbing the [cAMP](pm) rhythmicity. Knockdown of the IBMX-insensitive PDE8B by shRNA in MIN6 cells increased the basal level of [cAMP](pm) and prevented the [cAMP](pm)-lowering effect of 2',5'-dideoxyadenosine after exposure to IBMX. Moreover, PDE8B-knockdown cells showed reduced glucose-induced [cAMP](pm) oscillations and loss of the normal pulsatile pattern of insulin secretion. It is concluded that [cAMP](pm) oscillations in ß-cells are caused by periodic variations in cAMP generation, and that several PDEs, including PDE1, PDE3 and the IBMX-insensitive PDE8B, are required for shaping the sub-membrane cAMP signals and pulsatile insulin release.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/fisiologia , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/fisiologia , Insulina/metabolismo , Animais , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/fisiologia , Feminino , Glucose/fisiologia , Secreção de Insulina , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Isoenzimas/fisiologia , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Periodicidade , Fosfatos de Fosfatidilinositol/metabolismo , Sistemas do Segundo Mensageiro , Análise de Célula Única
16.
FASEB J ; 27(4): 1610-20, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23299857

RESUMO

Diacylglycerol (DAG) controls numerous cell functions by regulating the localization of C1-domain-containing proteins, including protein kinase C (PKC), but little is known about the spatiotemporal dynamics of the lipid. Here, we explored plasma membrane DAG dynamics in pancreatic ß cells and determined whether DAG signaling is involved in secretagogue-induced pulsatile release of insulin. Single MIN6 cells, primary mouse ß cells, and human ß cells within intact islets were transfected with translocation biosensors for DAG, PKC activity, or insulin secretion and imaged with total internal reflection fluorescence microscopy. Muscarinic receptor stimulation triggered stable, homogenous DAG elevations, whereas glucose induced short-lived (7.1 ± 0.4 s) but high-amplitude elevations (up to 109 ± 10% fluorescence increase) in spatially confined membrane regions. The spiking was mimicked by membrane depolarization and suppressed after inhibition of exocytosis or of purinergic P2Y1, but not P2X receptors, reflecting involvement of autocrine purinoceptor activation after exocytotic release of ATP. Each DAG spike caused local PKC activation with resulting dissociation of its substrate protein MARCKS from the plasma membrane. Inhibition of spiking reduced glucose-induced pulsatile insulin secretion. Thus, stimulus-specific DAG signaling patterns appear in the plasma membrane, including distinct microdomains, which have implications for the kinetic control of exocytosis and other membrane-associated processes.


Assuntos
Diglicerídeos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinase C/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Transdução de Sinais , Trifosfato de Adenosina/farmacologia , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Exocitose/efeitos dos fármacos , Glucose/metabolismo , Humanos , Secreção de Insulina , Camundongos , Transdução de Sinais/efeitos dos fármacos
17.
Domest Anim Endocrinol ; 87: 106836, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38141375

RESUMO

Feline diabetes mellitus is a common endocrine disease with increasing prevalence. It shows similarities with human type 2 diabetes and is characterized by insulin resistance and deficient insulin secretion. Moreover, cats and humans belong to the very few species that form amyloid depositions in the pancreatic islets. However, little is known about cat islet function and no studies have addressed insulin secretion from isolated islets ex vivo. The aim of this study was to establish a protocol for isolation of islets of Langerhans from pancreata of cats euthanized due to disease, and to evaluate insulin secretion responses to various physiological and pharmacological stimuli. Collagenase digestion of pancreatic tissue from 13 non-diabetic cats and two cats with diabetic ketoacidosis yielded individual islets surrounded by a layer of exocrine tissue that was reduced after two days in culture. Histological examination showed islet amyloid in pancreatic biopsies from most non-diabetic and in one diabetic cat. Islets from non-diabetic cats cultured at 5.5 mM glucose responded with increased insulin secretion to 16.7 mM glucose, 30 mM K+ and 20 µM of the sulfonylurea glipizide (2-3 times basal secretion at 3 mM glucose). The glucagon-like peptide-1 receptor agonist exendin-4 (100 nM) had no effect under basal conditions but potentiated glucose-triggered insulin release. Only one of nine islet batches from diabetic cats released detectable amounts of insulin, which was enhanced by exendin-4. Culture of islets from non-diabetic cats at 25 mM glucose impaired secretion both in response to glucose and K+ depolarization. In conclusion, we describe a procedure for isolation of islets from cat pancreas biopsies and demonstrate that isolated cat islets secrete insulin in response to glucose and antidiabetic drugs. The study provides a basis for future ex vivo studies of islet function relevant to the understanding of the pathophysiology and treatment of feline diabetes.


Assuntos
Doenças do Gato , Diabetes Mellitus Tipo 2 , Ilhotas Pancreáticas , Gatos , Animais , Humanos , Insulina/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/veterinária , Exenatida/farmacologia , Glucose/farmacologia , Doenças do Gato/tratamento farmacológico
18.
Cell Rep ; 43(4): 113992, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38536815

RESUMO

Insulin is packaged into secretory granules that depart the Golgi and undergo a maturation process that involves changes in the protein and lipid composition of the granules. Here, we show that insulin secretory granules form physical contacts with the endoplasmic reticulum and that the lipid exchange protein oxysterol-binding protein (OSBP) is recruited to these sites in a Ca2+-dependent manner. OSBP binding to insulin granules is positively regulated by phosphatidylinositol-4 (PI4)-kinases and negatively regulated by the PI4 phosphate (PI(4)P) phosphatase Sac2. Loss of Sac2 results in excess accumulation of cholesterol on insulin granules that is normalized when OSBP expression is reduced, and both acute inhibition and small interfering RNA (siRNA)-mediated knockdown of OSBP suppress glucose-stimulated insulin secretion without affecting insulin production or intracellular Ca2+ signaling. In conclusion, we show that lipid exchange at endoplasmic reticulum (ER)-granule contact sites is involved in the exocytic process and propose that these contacts act as reaction centers with multimodal functions during insulin granule maturation.


Assuntos
Colesterol , Retículo Endoplasmático , Secreção de Insulina , Insulina , Antígenos de Histocompatibilidade Menor , Receptores de Esteroides , Vesículas Secretórias , Retículo Endoplasmático/metabolismo , Vesículas Secretórias/metabolismo , Animais , Colesterol/metabolismo , Insulina/metabolismo , Receptores de Esteroides/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Camundongos , Humanos , Cálcio/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Glucose/metabolismo
19.
J Biol Chem ; 287(13): 9862-9872, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22298778

RESUMO

The events leading to the activation of store-operated Ca(2+) entry (SOCE) involve Ca(2+) depletion of the endoplasmic reticulum (ER) resulting in translocation of the transmembrane Ca(2+) sensor protein, stromal interaction molecule 1 (STIM1), to the junctions between ER and the plasma membrane where it binds to the Ca(2+) channel protein Orai1 to activate Ca(2+) influx. Using confocal and total internal reflection fluorescence microscopy, we studied redistribution kinetics of fluorescence-tagged STIM1 and Orai1 as well as SOCE in insulin-releasing ß-cells and glucagon-secreting α-cells within intact mouse and human pancreatic islets. ER Ca(2+) depletion triggered accumulation of STIM1 puncta in the subplasmalemmal ER where they co-clustered with Orai1 in the plasma membrane and activated SOCE. Glucose, which promotes Ca(2+) store filling and inhibits SOCE, stimulated retranslocation of STIM1 to the bulk ER. This effect was evident at much lower glucose concentrations in α- than in ß-cells consistent with involvement of SOCE in the regulation of glucagon secretion. Epinephrine stimulated subplasmalemmal translocation of STIM1 in α-cells and retranslocation in ß-cells involving raising and lowering of cAMP, respectively. The cAMP effect was mediated both by protein kinase A and exchange protein directly activated by cAMP. However, the cAMP-induced STIM1 puncta did not co-cluster with Orai1, and there was no activation of SOCE. STIM1 translocation can consequently occur independently of Orai1 clustering and SOCE.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Adulto , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Retículo Endoplasmático/metabolismo , Epinefrina/farmacologia , Feminino , Células Secretoras de Glucagon/citologia , Humanos , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Pessoa de Meia-Idade , Proteína ORAI1 , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Molécula 1 de Interação Estromal
20.
Cell Signal ; 109: 110805, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37437828

RESUMO

Genetically encoded Ca2+ indicators have become widely used in cell signalling studies as they offer advantages over cell-loaded dye indicators in enabling specific cellular or subcellular targeting. Comparing responses from dye and protein-based indicators may provide information about indicator properties and cell physiology, but side-by-side recordings in cells are scarce. In this study, we compared cytoplasmic Ca2+ concentration ([Ca2+]i) changes in insulin-secreting ß-cells recorded with commonly used dyes and indicators based on circularly permuted fluorescent proteins. Total internal reflection fluorescence (TIRF) imaging of K+ depolarization-triggered submembrane [Ca2+]i increases showed that the dyes Fluo-4 and Fluo-5F mainly reported stable [Ca2+]i elevations, whereas the proteins R-GECO1 and GCaMP5G more often reported distinct [Ca2+]i spikes from an elevated level. [Ca2+]i spiking occurred also in glucose-stimulated cells. The spikes reflected Ca2+ release from the endoplasmic reticulum, triggered by autocrine activation of purinergic receptors after exocytotic release of ATP and/or ADP, and the spikes were consequently prevented by SERCA inhibition or P2Y1-receptor antagonism. Widefield imaging, which monitors the entire cytoplasm, increased the spike detection by the Ca2+ dyes. The indicator-dependent response patterns were unrelated to Ca2+ binding affinity, buffering and mobility, and probably reflects the much slower dissociation kinetics of protein compared to dye indicators. Ca2+ dyes thus report signalling within the submembrane space excited by TIRF illumination, whereas the protein indicators also catch Ca2+ events originating outside this volume. The study highlights that voltage-dependent Ca2+ entry in ß-cells is tightly linked to local intracellular Ca2+ release mediated via an autocrine route that may be more important than previously reported direct Ca2+ effects on phospholipase C or on intracellular channels mediating calcium-induced calcium release.


Assuntos
Cálcio , Células Secretoras de Insulina , Cálcio/metabolismo , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Retículo Endoplasmático/metabolismo , Corantes/metabolismo , Corantes/farmacologia , Sinalização do Cálcio , Trifosfato de Adenosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA