Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 77(22): 4709-4723, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31925452

RESUMEN

AIMS: Endocannabinoids are lipid mediators involved in the regulation of glucose homeostasis. They interact with the canonical cannabinoid receptors CB1 and CB2, and it is now apparent that some cannabinoid receptor ligands are also agonists at GPR55. Thus, CB1 antagonists such as SR141716A, also known as rimonabant, and AM251 act as GPR55 agonists in some cell types. The complex pharmacological properties of cannabinoids make it difficult to fully identify the relative importance of CB1 and GPR55 in the functional effects of SR141716A, and AM251. Here, we determine whether SR141716A and AM251 regulation of mouse and human islet function is through their action as GPR55 agonists. METHODS: Islets isolated from Gpr55+/+ and Gpr55-/- mice and human donors were incubated in the absence or presence of 10 µM SR141716A or AM251, concentrations that are known to activate GPR55. Insulin secretion, cAMP, IP1, apoptosis and ß-cell proliferation were quantified by standard techniques. RESULTS: Our results provide the first evidence that SR141716A and AM251 are not GPR55 agonists in islets, as their effects are maintained in islets isolated from Gpr55-/- mice. Their signalling through Gq-coupled cascades to induce insulin secretion and human ß-cell proliferation, and protect against apoptosis in vitro, indicate that they have direct beneficial effects on islet function. CONCLUSION: These observations may be useful in directing development of peripherally restricted novel therapeutics that are structurally related to SR141716A and AM251, and which potentiate glucose-induced insulin secretion and stimulate ß-cell proliferation.


Asunto(s)
Cannabinoides/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , Receptores de Cannabinoides/metabolismo , Rimonabant/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Endocannabinoides/metabolismo , Femenino , Humanos , Insulina , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/metabolismo
2.
Cell Mol Life Sci ; 75(4): 715-726, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28921001

RESUMEN

AIMS: Complement components 3 and 5 (C3 and C5) play essential roles in the complement system, generating C3a and C5a peptides that are best known as chemotactic and inflammatory factors. In this study we characterised islet expression of C3 and C5 complement components, and the impact of C3aR and C5aR1 activation on islet function and viability. MATERIALS AND METHODS: Human and mouse islet mRNAs encoding key elements of the complement system were quantified by qPCR and distribution of C3 and C5 proteins was determined by immunohistochemistry. Activation of C3aR and C5aR1 was determined using DiscoverX beta-arrestin assays. Insulin secretion from human and mouse islets was measured by radioimmunoassay, and intracellular calcium ([Ca2+]i), ATP generation and apoptosis were assessed by standard techniques. RESULTS: C3 and C5 proteins and C3aR and C5aR1 were expressed by human and mouse islets, and C3 and C5 were mainly localised to ß- and α-cells. Conditioned media from islets exposed for 1 h to 5.5 and 20 mM glucose stimulated C3aR and C5aR1-driven beta-arrestin recruitment. Activation of C3aR and C5aR1 potentiated glucose-induced insulin secretion from human and mouse islets, increased [Ca2+]i and ATP generation, and protected islets against apoptosis induced by a pro-apoptotic cytokine cocktail or palmitate. CONCLUSIONS: Our observations demonstrate a functional link between activation of components of the innate immune system and improved ß-cell function, suggesting that low-level chronic inflammation may improve glucose homeostasis through direct effects on ß-cells.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Complemento/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Complemento C3/metabolismo , Complemento C5/metabolismo , Citocinas/metabolismo , Glucosa/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , beta-Arrestinas/metabolismo
3.
Cell Mol Life Sci ; 75(16): 3039-3050, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29455414

RESUMEN

INTRODUCTION: Islets synthesise and secrete numerous peptides, some of which are known to be important regulators of islet function and glucose homeostasis. In this study, we quantified mRNAs encoding all peptide ligands of islet G protein-coupled receptors (GPCRs) in isolated human and mouse islets and carried out in vitro islet hormone secretion studies to provide functional confirmation for the species-specific role of peptide YY (PYY) in mouse islets. MATERIALS AND METHODS: GPCR peptide ligand mRNAs in human and mouse islets were quantified by quantitative real-time PCR relative to the reference genes ACTB, GAPDH, PPIA, TBP and TFRC. The pathways connecting GPCR peptide ligands with their receptors were identified by manual searches in the PubMed, IUPHAR and Ingenuity databases. Distribution of PYY protein in mouse and human islets was determined by immunohistochemistry. Insulin, glucagon and somatostatin secretion from islets was measured by radioimmunoassay. RESULTS: We have quantified GPCR peptide ligand mRNA expression in human and mouse islets and created specific signalomes mapping the pathways by which islet peptide ligands regulate human and mouse GPCR signalling. We also identified species-specific islet expression of several GPCR ligands. In particular, PYY mRNA levels were ~ 40,000-fold higher in mouse than human islets, suggesting a more important role of locally secreted Pyy in mouse islets. This was confirmed by IHC and functional experiments measuring insulin, glucagon and somatostatin secretion. DISCUSSION: The detailed human and mouse islet GPCR peptide ligand atlases will allow accurate translation of mouse islet functional studies for the identification of GPCR/peptide signalling pathways relevant for human physiology, which may lead to novel treatment modalities of diabetes and metabolic disease.


Asunto(s)
Islotes Pancreáticos/metabolismo , Péptido YY/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Expresión Génica , Humanos , Inmunohistoquímica , Ligandos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Péptido YY/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Diabetes Obes Metab ; 20(3): 599-609, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28940946

RESUMEN

AIMS: Two unmet therapeutic strategies for diabetes treatment are prevention of beta-cell death and stimulation of beta-cell replication. Our aim was to characterize the role of neuropeptide Y receptors in the control of beta-cell mass. MATERIALS AND METHODS: We used endogenous and selective agonists of the NPY receptor system to explore its role in the prevention of beta-cell apoptosis and proliferation in islets isolated from both mouse and human donors. We further explored the intra-cellular signalling cascades involved, using chemical inhibitors of key signalling pathways. As proof of principle we designed a long-acting analogue of [Leu31 Pro34 ]-NPY, an agonist of the islet-expressed Y receptors, to determine if targeting this system could preserve beta-cell mass in vivo. RESULTS: Our data reveal that NPY Y1, 4 and 5 receptor activation engages a generalized and powerful anti-apoptotic pathway that protects mouse and human islets from damage. These anti-apoptotic effects were dependent on stimulating a Gαi-PLC-PKC signalling cascade, which prevented cytokine-induced NFkB signalling. NPY receptor activation functionally protected islets by restoring glucose responsiveness following chemically induced injury in both species. NPY receptor activation attenuated beta-cell apoptosis, preserved functional beta-cell mass and attenuated the hyperglycaemic phenotype in a low-dose streptozotocin model of diabetes. CONCLUSION: Taken together, our observations identify the islet Y receptors as promising targets for the preservation of beta-cell mass. As such, targeting these receptors could help to maintain beta-cell mass in both type 1 and type 2 diabetes, and may also be useful for improving islet transplantation outcomes.


Asunto(s)
Células Secretoras de Insulina/citología , Receptores de Neuropéptido Y/fisiología , Análisis de Varianza , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Humanos , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal/fisiología
5.
Diabetes Obes Metab ; 20(4): 930-942, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29205751

RESUMEN

AIMS: To examine the effects of Abn-CBD (GPR55 agonist) and LH-21 (CB1 antagonist) on human and mouse islet function, and to determine signalling via GPR55 using islets from GPR55-/- mice. MATERIALS AND METHODS: Islets isolated from human organ donors and mice were incubated in the absence or presence of Abn-CBD or LH-21, and insulin secretion, [Ca2+ ]i, cAMP, apoptosis, ß-cell proliferation and CREB and AKT phosphorylation were examined using standard techniques. RESULTS: Abn-CBD potentiated glucose-stimulated insulin secretion and elevated [Ca2+ ]i in human islets and islets from both GPR55+/+ and GPR55-/- mice. LH-21 also increased insulin secretion and [Ca2+ ]i in human islets and GPR55+/+ mouse islets, but concentrations of LH-21 up to 0.1 µM were ineffective in islets from GPR55-/- mice. Neither ligand affected basal insulin secretion or islet cAMP levels. Abn-CBD and LH-21 reduced cytokine-induced apoptosis in human islets and GPR55+/+ mouse islets, and these effects were suppressed after GPR55 deletion. They also increased ß-cell proliferation: the effects of Abn-CBD were preserved in islets from GPR55-/- mice, while those of LH-21 were abolished. Abn-CBD and LH-21 increased AKT phosphorylation in mouse and human islets. CONCLUSIONS: This study showed that Abn-CBD and LH-21 improve human and mouse islet ß-cell function and viability. Use of islets from GPR55-/- mice suggests that designation of Abn-CBD and LH-21 as a GPR55 agonist and a CB1 antagonist, should be revised.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Resorcinoles/farmacología , Triazoles/farmacología , Adulto , Animales , Células Cultivadas , Femenino , Humanos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Cannabinoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Cell Physiol Biochem ; 44(4): 1352-1359, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29186709

RESUMEN

BACKGROUND/AIMS: Rodent islets are often used for basic science research but they do not always recapitulate signalling events in human islets. This study evaluated the glucose-dependent responses of human and mouse islets in terms of dynamic insulin secretion, metabolic coupling and the role of glucose transporters. METHODS: Glucose-induced insulin secretion from isolated mouse and human islets was profiled by perifusion and islet ATP levels were measured by chemoluminescence assay. Glucose transporter expression was determined by qPCR and western blotting. RESULTS: Human islets show a left-shifted glucose concentration-insulin secretion profile compared to mouse islets. These data are consistent with glucose transporter expression, with human islets expressing mainly GLUT1 and GLUT3, and GLUT2 being the predominant transporter in mouse islets. Using the GLUT1 inhibitor STF-31 we unveiled an important role for GLUT1 for differences in glucose-induced insulin secretion profiles observed between the two species. CONCLUSION: The high affinity of GLUT1/3 for glucose reflects the left-shifted glucose-induced insulin secretory response of human islets and the impairment of insulin secretion from human islets after STF-31 treatment indicates an important role for GLUT1 in human islet stimulus-secretion coupling. Our data provide further insight into key differences between insulin secretion regulation in mouse and human islets.


Asunto(s)
Adenosina Trifosfato/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Adulto , Animales , Femenino , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 2/antagonistas & inhibidores , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Transportador de Glucosa de Tipo 3/antagonistas & inhibidores , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Humanos , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Cinética , Masculino , Ratones , Persona de Mediana Edad , Piridinas/farmacología , ARN Mensajero/metabolismo
7.
Clin Sci (Lond) ; 131(23): 2835-2845, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101297

RESUMEN

AIMS: The aims of the present study were (i) to determine whether the reported beneficial effects of mesenchymal stromal cells (MSCs) on mouse islet function extend to clinically relevant human tissues (islets and MSCs), enabling translation into improved protocols for clinical human islet transplantation; and (ii) to identify possible mechanisms through which human MSCs influence human islet function. MATERIALS AND METHODS: Human islets were co-cultured with human adipose tissue-derived MSCs (hASCs) or pre-treated with its products - extracellular matrix (ECM) and annexin A1 (ANXA1). Mouse islets were pre-treated with mouse MSC-derived ECM. Islet insulin secretory function was assessed in vitro by radioimmunoassay. Quantitative RT-PCR was used to screen human adipMSCs for potential ligands of human islet G-protein-coupled receptors. RESULTS: We show that co-culture with hASCs improves human islet secretory function in vitro, as measured by glucose-stimulated insulin secretion, confirming previous reports using rodent tissues. Furthermore, we demonstrate that these beneficial effects on islet function can be partly attributed to the MSC-derived products ECM and ANXA1. CONCLUSIONS: Our results suggest that hASCs have the potential to improve the quality of human islets isolated for transplantation therapy of Type 1 diabetes. Furthermore, it may be possible to achieve improvements in human islet quality in a cell-free culture system by using the MSC-derived products ANXA1 and ECM.


Asunto(s)
Matriz Extracelular/fisiología , Islotes Pancreáticos/fisiología , Células Madre Mesenquimatosas/fisiología , Tejido Adiposo/citología , Animales , Anexina A1/metabolismo , Anexina A1/farmacología , Técnicas de Cocultivo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ligandos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Receptores Odorantes/metabolismo
8.
Diabetologia ; 56(11): 2467-76, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979485

RESUMEN

AIMS/HYPOTHESIS: Chemokine (C-C motif) ligand 5 (CCL5) acts at C-C chemokine receptors (CCRs) to promote immune cell recruitment to sites of inflammation, but is also an agonist at G-protein-coupled receptor 75 (GPR75), which has very limited homology with CCRs. GPR75 is coupled to Gq to elevate intracellular calcium, so we investigated whether islets express this receptor and whether its activation by CCL5 increases beta cell calcium levels and insulin secretion. METHODS: Islet CCL5 receptor mRNA expression was measured by quantitative RT-PCR and GPR75 was detected in islets by western blotting and immunohistochemistry. In some experiments GPR75 was downregulated by transient transfection with small interfering RNA. Real-time changes in intracellular calcium were determined by single-cell microfluorimetry. Dynamic insulin secretion from perifused islets was quantified by radioimmunoassay. Glucose homeostasis in lean and obese mice was determined by measuring glucose and insulin tolerance, and insulin secretion in vivo. RESULTS: Mouse and human islets express GPR75 and its ligand CCL5. Exogenous CCL5 reversibly increased intracellular calcium in beta cells via GPR75, this phenomenon being dependent on phospholipase C activation and calcium influx. CCL5 also stimulated insulin secretion from mouse and human islets in vitro, and improved glucose tolerance in lean mice and in a mouse model of hyperglycaemia and insulin resistance (ob/ob). The improvement in glucose tolerance was associated with enhanced insulin secretion in vivo, without changes in insulin sensitivity. CONCLUSIONS/INTERPRETATION: Although CCL5 is implicated in the pathogenesis of diabetes through activation of CCRs, it has beneficial effects on beta cells through GPR75 activation.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiocina CCL5/metabolismo , Homeostasis/genética , Homeostasis/fisiología , Humanos , Inmunohistoquímica , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos ICR , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Ethnopharmacol ; 302(Pt B): 115937, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36410575

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ayurvedic medicine has been used in the treatment of diabetes mellitus for centuries. In Arabia and some areas of Africa, Commiphora myrrha (CM) has been extensively used as a plant-based remedy. We have previously shown that an aqueous CM resin solution directly stimulates insulin secretion from MIN6 cells, a mouse ß-cell line, and isolated mouse and human islets. However, the signaling pathways involved in CM-induced insulin secretion are completely unknown. Insulin secretion is normally triggered by elevations in intracellular Ca2+ ([Ca2+]i) through voltage gated Ca2+ channels (VGCC) and activation of protein kinases. Protein and lipid kinases such as protein kinase A (PKA), Ca2+-calmodulin dependent protein kinase II (CaMKII), phosphoinositide 3-kinases (PI3Ks), protein kinase C (PKC) and mitogen-activated protein kinase (MAPK), specifically extracellular signal-regulated kinases (ERK1/2), may be involved in receptor-operated insulin secretion. Therefore, we hypothesized that CM may induce insulin secretion by modulating the activity of VGCC and/or one or more of the above kinases. AIM OF THE STUDY: To investigate the possible molecular mechanism of action of CM-induced insulin secretion. The effects of aqueous CM resin extract on [Ca2+]i and protein kinase activation from ß-cells were examined. METHODS: The effect of aqueous CM resin solution on [Ca2+]i was assessed using Ca2+ microfluorimetry. The involvement of VGCC in CM-induced insulin secretion was investigated using static and perifusion insulin secretion experiments in the presence of either EGTA, a Ca2+ chelator, or nifedipine, a blocker of VGCC. The involvement of kinase activation in the stimulatory effect of CM on insulin secretion was examined by using static and perifusion insulin secretion experiments in the presence of known pharmacological inhibitors and/or downregulation of specific kinases. The effects of CM on phosphorylation of PKCζ and ERK1/2 were also assessed using the Wes™ capillary-based protein electrophoresis. RESULTS: Ca2+ microfluorimetry measurements showed that exposing MIN6 cells to CM (0.5-2 mg/mL) was not associated with changes in [Ca2+]i. Similarly, incubating MIN6 cells and mouse islets with EGTA and nifedipine, respectively, did not attenuate the insulin secretion induced by CM. However, incubating mouse and human islets with CM in the presence of staurosporine, a non-selective protein kinase inhibitor, completely blocked the effect of CM on insulin secretion. Exposing mouse islets to CM in the presence of H89, KN62 and LY294002, inhibitors of PKA, CaMKII and PI3K, respectively, did not reduce CM-induced insulin secretion. However, incubating mouse and human islets with CM in the presence of Ro 31-8220, a pan-PKC inhibitor, diminished insulin secretion stimulated by CM, whereas inhibiting the action of typical PKC (with Go6976) and PLCß (with U73122) did not affect CM-stimulated insulin secretion. Similarly, downregulating typical and novel PKC by chronic exposure of mouse islets to phorbol 12-myristate 13-acetate (PMA) was also not associated with a decrease in the stimulatory effect of CM on insulin secretion. Interestingly, CM-induced insulin secretion from mouse islets was inhibited in the presence of the PKCζ inhibitor ZIP and a MAPK inhibitor PD 98059. In addition, Wes™ capillary-based protein electrophoresis indicated that expression of the phosphorylated forms of PKCζ and ERK1/2, a MAPK, was significantly increased following exposure of INS-1832/13 cells, a rat insulinoma cell line, to CM. CONCLUSIONS: Our data indicate that CM directly stimulates insulin secretion through activating known downstream effectors of insulin-stimulus secretion coupling. Indeed, the increase in insulin secretion seen with CM is independent of changes in [Ca2+]i and does not involve activation of VGCC. Instead, the CM stimulatory effect on insulin secretion is completely dependent on protein kinase activation. Our findings indicate that CM could induce insulin exocytosis by stimulating the phosphorylation and activation of PKCζ, which in turn phosphorylates and activates ERK1/2.


Asunto(s)
Commiphora , Neoplasias Pancreáticas , Humanos , Ratas , Animales , Ratones , Secreción de Insulina , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Ácido Egtácico , Nifedipino , Proteína Quinasa C , Proteínas Quinasas Dependientes de AMP Cíclico , Insulina , Quinasas MAP Reguladas por Señal Extracelular , Acetato de Tetradecanoilforbol , Fosfatidilinositol 3-Quinasas
10.
Sci Rep ; 12(1): 9383, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672347

RESUMEN

Neuromedin B (NMB) is a member of the neuromedin family of neuropeptides with a high level of region-specific expression in the brain. Several GWAS studies on non-obese and obese patients suggested that polymorphisms in NMB predispose to obesity by affecting appetite control and feeding preference. Furthermore, several studies proposed that NMB can act as an insulin releasing peptide. Since the functional study has never been done, the in vivo role of NMB as modulator of weight gain or glucose metabolism remains unclear. Here, we generated Nmb conditional mice and nervous system deficient NmB mice. We then performed olfactory and food preference analysis, as well as metabolic analysis under standard and high fat diet. Additionally, in direct islet studies we evaluated the role of NMB on basal and glucose-stimulated insulin secretion in mouse and humans.


Asunto(s)
Insulina , Neuropéptidos , Animales , Peso Corporal , Glucosa , Homeostasis , Humanos , Insulina/metabolismo , Ratones , Neuroquinina B/análogos & derivados , Neuroquinina B/metabolismo , Neuropéptidos/genética , Obesidad/genética
11.
Metabolism ; 134: 155247, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35760117

RESUMEN

AIMS: Hypertriglyceridemia is associated with an increased risk of type 2 diabetes. We aimed to comprehensively examine the effects of hypertriglyceridemia on major glucose homeostatic mechanisms involved in diabetes progression. METHODS: In this randomized, cross-over, single-blinded study, two dual-labeled, 3-hour oral glucose tolerance tests were performed during 5-hour intravenous infusions of either 20 % Intralipid or saline in 12 healthy subjects (age 27.9 ± 2.6 years, 11 men, BMI 22.6 ± 1.4 kg/m2) to evaluate lipid-induced changes in insulin metabolism and glucose kinetics. Insulin sensitivity, ß cell secretory function, and insulin clearance were assessed by modeling glucose, insulin and C-peptide data. Intestinal glucose absorption, endogenous glucose production, and glucose clearance were assessed from glucose tracers. The effect of triglycerides on ß-cell secretory function was examined in perifusion experiments in murine pseudoislets and human pancreatic islets. RESULTS: Mild acute hypertriglyceridemia impaired oral glucose tolerance (mean glucose: +0.9 [0.3, 1.5] mmol/L, p = 0.008) and whole-body insulin sensitivity (Matsuda index: -1.67 [-0.50, -2.84], p = 0.009). Post-glucose hyperinsulinemia (mean insulin: +99 [17, 182] pmol/L, p = 0.009) resulted from reduced insulin clearance (-0.16 [-0.32, -0.01] L min-1 m-2, p = 0.04) and enhanced hyperglycemia-induced total insulin secretion (+11.9 [1.1, 22.8] nmol/m2, p = 0.02), which occurred despite a decline in model-derived ß cell glucose sensitivity (-41 [-74, -7] pmol min-1 m-2 mmol-1 L, p = 0.04). The analysis of tracer-derived glucose metabolic fluxes during lipid infusion revealed lower glucose clearance (-96 [-152, -41] mL/kgFFM, p = 0.005), increased 2-hour oral glucose absorption (+380 [42, 718] µmol/kgFFM, p = 0.04) and suppressed endogenous glucose production (-448 [-573, -123] µmol/kgFFM, p = 0.005). High-physiologic triglyceride levels increased acute basal insulin secretion in murine pseudoislets (+11 [3, 19] pg/aliquot, p = 0.02) and human pancreatic islets (+286 [59, 512] pg/islet, p = 0.02). CONCLUSION: Our findings support a critical role for hypertriglyceridemia in the pathogenesis of type 2 diabetes in otherwise healthy individuals and dissect the glucose homeostatic mechanisms involved, encompassing insulin sensitivity, ß cell function and oral glucose absorption.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Hipertrigliceridemia , Resistencia a la Insulina , Adulto , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Humanos , Insulina/metabolismo , Cinética , Masculino , Ratones , Triglicéridos
12.
J Clin Invest ; 118(10): 3390-402, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802479

RESUMEN

The final pathway of beta cell destruction leading to insulin deficiency, hyperglycemia, and clinical type 1 diabetes is unknown. Here we show that circulating CTLs can kill beta cells via recognition of a glucose-regulated epitope. First, we identified 2 naturally processed epitopes from the human preproinsulin signal peptide by elution from HLA-A2 (specifically, the protein encoded by the A*0201 allele) molecules. Processing of these was unconventional, requiring neither the proteasome nor transporter associated with processing (TAP). However, both epitopes were major targets for circulating effector CD8+ T cells from HLA-A2+ patients with type 1 diabetes. Moreover, cloned preproinsulin signal peptide-specific CD8+ T cells killed human beta cells in vitro. Critically, at high glucose concentration, beta cell presentation of preproinsulin signal epitope increased, as did CTL killing. This study provides direct evidence that autoreactive CTLs are present in the circulation of patients with type 1 diabetes and that they can kill human beta cells. These results also identify a mechanism of self-antigen presentation that is under pathophysiological regulation and could expose insulin-producing beta cells to increasing cytotoxicity at the later stages of the development of clinical diabetes. Our findings suggest that autoreactive CTLs are important targets for immune-based interventions in type 1 diabetes and argue for early, aggressive insulin therapy to preserve remaining beta cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Epítopos de Linfocito T/inmunología , Glucosa/metabolismo , Células Secretoras de Insulina/patología , Insulina/inmunología , Precursores de Proteínas/inmunología , Adolescente , Adulto , Epítopos de Linfocito T/efectos de los fármacos , Femenino , Glucosa/farmacología , Humanos , Insulina/metabolismo , Células K562 , Masculino , Fenotipo , Precursores de Proteínas/metabolismo , Señales de Clasificación de Proteína
13.
J Ethnopharmacol ; 264: 113075, 2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-32829055

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Traditionally plant-based remedies such as Commiphora myrrha (CM) have been used as an ayurvedic medicine to treat diabetes mellitus in some region of Arabia and Africa. Previous reports have shown that CM reduced blood glucose levels and increased insulin concentrations in animal models of diabetes in vivo. However, the exact mechanisms by which CM improved glycemic control in these animals are not fully understood. We hypothesized that CM may have a direct insulinotropic activity on ß-cells to increase insulin secretion. AIM OF THE STUDY: The direct effects of CM were investigated using MIN6 ß-cells and isolated mouse and human islets in static and perifusion insulin secretion experiments. Isolated mouse and human islets were used to investigate the rate and pattern of CM-induced insulin secretion. MATERIALS AND METHODS: The effect of CM on insulin secretion was assessed by static and perifusion experiments using MIN6 cells, a mouse-derived ß-cell line, and primary mouse and human islets. The effects of CM on cell viability and membrane integrity of MIN6 cells and mouse islets were assessed using an ATP viability assay and a trypan blue exclusion test. The mRNA expression profiles of preproinsulin and Pdx1, a major ß-cell transcription factor, were determined by quantitative RT-PCR following chronic exposure to CM. RESULTS: Exposing MIN6 cells to a CM resin solution (0.5-10 mg/ml) caused a concentration-dependent increase in insulin secretion in a static setting. Similarly, incubating mouse islets to CM (0.1-10 mg/ml) resulted in stimulation of insulin secretion in a concentration-dependent manner. CM concentrations at ≤ 2 mg/ml were not associated with reduction in cell viability nor with reduction in cell membrane integrity. However, higher concentrations of CM were accompanied with marked uptake of trypan blue dye and cell death. In a perifusion setting, CM (2 mg/ml) caused rapid and reversible increases in insulin secretion from both mouse and human islets at both sub-stimulatory and stimulatory glucose levels. The stimulatory effect of CM on insulin secretion did not change the total insulin content of ß-cells nor the mRNA expression of preproinsulin and Pdx1. CONCLUSIONS: These data indicate that aqueous CM resin solution has a direct stimulatory effect on ß-cells without compromising plasma membrane integrity. CM stimulates insulin secretion from MIN6 cells, a mouse-derived ß-cell line, and isolated primary mouse and human islets in vitro at both sub-stimulatory and stimulatory glucose concentrations. The mechanism by which CM may induce insulin secretion is most likely due to a stimulation of insulin granules release rather than insulin synthesis.


Asunto(s)
Commiphora , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Extractos Vegetales/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Secreción de Insulina/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/aislamiento & purificación
14.
Cells ; 10(3)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33809893

RESUMEN

AIMS: The endocannabinoid system is a complex cell-signaling network through which endogenous cannabinoid ligands regulate cell function by interaction with CB1 and CB2 cannabinoid receptors, and with the novel cannabinoid receptor GPR55. CB1, CB2, and GPR55 are expressed by islet ß-cells where they modulate insulin secretion. We have previously shown that administration of the putative CB2 antagonist/inverse agonist JTE 907 to human islets did not affect the insulinotropic actions of CB2 agonists and it unexpectedly stimulated insulin secretion on its own. In this study, we evaluated whether the lack of antagonism could be related to the ability of JTE 907 to act as a GPR55 agonist. MATERIALS AND METHODS: We used islets isolated from human donors and from Gpr55+/+ and Gpr55-/- mice and quantified the effects of incubation with 10 µM JTE 907 on dynamic insulin secretion, apoptosis, and ß-cell proliferation by radioimmunoassay, luminescence caspase 3/7 activity, and immunofluorescence, respectively. We also measured islet IP1 and cAMP accumulation using fluorescence assays, and monitored [Ca2+]i elevations by Fura-2 single cell microfluorometry. RESULTS: JTE 907 significantly stimulated insulin secretion from islets isolated from human donors and islets from Gpr55+/+ and Gpr55-/- mice. These stimulatory effects were accompanied by significant elevations of IP1 and [Ca2+]i, but there were no changes in cAMP generation. JTE 907 also significantly reduced cytokine-induced apoptosis in human and mouse islets and promoted human ß-cell proliferation. CONCLUSION: Our observations show for the first time that JTE 907 acts as a Gq-coupled agonist in islets to stimulate insulin secretion and maintain ß-cell mass in a GPR55-independent fashion.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Dioxoles/farmacología , Islotes Pancreáticos/efectos de los fármacos , Quinolonas/farmacología , Receptores de Cannabinoides/efectos de los fármacos , Adulto , Animales , Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Fosfatos de Inositol/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ligandos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo , Técnicas de Cultivo de Tejidos
15.
Cell Physiol Biochem ; 26(6): 1051-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21220936

RESUMEN

Plant-derived extracts have been used as folk remedies for Type 2 diabetes mellitus (T2DM) for many centuries, and offer the potential of cheap and readily available alternatives to conventional pharmaceuticals in developing countries. Extracts of Costus pictus (CP), a plant belonging to the Costaceae family, are reported to have antidiabetic activity in vivo. The exact molecular mode of action(s) of CP is unclear but the antihyperglycemic effect seen in animal studies was associated with dramatic increases in insulin secretion so in our study we have measured the effect of aqueous CP extract on insulin secretion in vitro from the MIN6 ß-cell line and isolated mouse and human islets. Our data demonstrate that CP has a direct stimulatory effect on insulin secretion at basal but not stimulatory glucose concentrations which was not associated with compromised membrane integrity or decrease ß-cell viability. Single cell calcium microfluorimetry measurements showed that CP caused elevations in ß-cell intracellular Ca(2+) concentrations ([Ca(2+)](i)), an effect which was completely abolished by the removal of extracellular Ca(2+) or blockade of voltage-gated Ca(2+) channels (VGCC). These in vitro observations suggest that one mode of action of CP is through stimulating insulin secretion which may be mediated, in part, by the ability of CP to increase [Ca(2+)](i) levels through VGCC. CP extracts may provide an affordable and inexpensive alternative for treating patients with T2DM.


Asunto(s)
Canales de Calcio/química , Costus/química , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Línea Celular , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Extractos Vegetales/química
16.
Metabolism ; 113: 154394, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33058852

RESUMEN

BACKGROUND AND PURPOSE: Chemokines are known to play essential roles mediating immunity and inflammation in many physiological and pathophysiological processes, with reports linking their action to the development of obesity, insulin resistance and type 2 diabetes (T2D). Given our findings of highly upregulated mRNA expression of the chemokine receptor CCR9 in islets from obese human donors, we have determined the effects of CCR9 activation by CCL25 on islet function and viability. BASIC PROCEDURES: RT-qPCR was used to measure expression of 384 GPCR mRNAs in human islets from organ donors with normal and elevated BMI. mRNA encoding CCR9, a receptor that was highly upregulated in islets from obese donors, was also quantified in islets from lean and high-fat diet (HFD) mice. The effects of CCR9 activation by exogenous CCL25 in human and mouse islets and its inhibition by the CCR9 antagonist vercirnon on insulin secretion, apoptosis and cAMP accumulation were examined using standard techniques. MAIN FINDINGS: The qPCR analysis showed altered expression of several GPCRs in islets isolated from lean and obese donors. CCR9 displayed over 90-fold upregulation in islets from obese individuals, and it was also significantly upregulated in islets from obese mice. In isolated human and mouse islets exogenous CCL25 inhibited glucose-induced insulin secretion in a concentration-dependent manner, enhanced cytokine-induced apoptosis and significantly reduced forskolin-induced elevation in cAMP levels. These detrimental effects of CCL25 in islets were blocked by vercirnon, which had no effect on its own. PRINCIPAL CONCLUSIONS: We have shown that CCL25 acts via the Gαi-coupled receptor CCR9 to impair beta-cell function by inhibiting insulin secretion and promoting cytokine-induced apoptosis. Upregulation of CCR9 in islets in obesity, possibly secondary to accumulation of passenger immune cells, may predispose to metabolic dysfunction and our data suggest that CCL25 downregulation or CCR9 inhibition could be explored to treat T2D.


Asunto(s)
Quimiocinas CC/fisiología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Secretoras de Insulina/patología , Receptores CCR/fisiología , Animales , Índice de Masa Corporal , Quimiocinas CC/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Hipoglucemiantes/uso terapéutico , Ratones , Receptores CCR/metabolismo
17.
Stem Cells Transl Med ; 8(9): 935-944, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31066521

RESUMEN

Islet transplantation has the potential to cure type 1 diabetes, but current transplantation protocols are not optimal and there is extensive loss of islet ß-cell insulin secretory function during the immediate post-transplantation period. Studies using experimental models of diabetes have shown that the coculture of islets with mesenchymal stromal cells (MSCs) prior to transplantation improves graft function, but several variables differed among research groups (e.g., type of MSCs used and the treatment conditions). We have therefore assessed the effects of MSCs on mouse and human islets by investigating the importance of tissue source for MSCs, the coculture protocol configuration and length, the effect of activated MSCs, and different ß-cell secretory stimuli. MSCs derived from adipose tissue (aMSCs) were the most effective at supporting ß-cell insulin secretion in both mouse and human islets, in a direct contact coculture configuration. Preculture with aMSCs enhanced both phases of glucose-induced insulin secretion and further enhanced secretory responses to the non-nutrients carbachol and arginine. These effects required a coculture period of 48-72 hours and were not dependent on activation of the MSCs. Thus, direct contact coculture with autologous, adipose-derived MSCs for a minimum of 48 hours before implantation is likely to be an effective addition to human islet transplantation protocols. Stem Cells Translational Medicine 2019;8:935&944.


Asunto(s)
Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Tejido Adiposo/citología , Animales , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Técnicas de Cocultivo , Glucosa/farmacología , Humanos , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Páncreas/citología , Factor de Necrosis Tumoral alfa/farmacología
18.
Diabetes ; 56(1): 197-203, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17192482

RESUMEN

The roles played by arachidonic acid and its cyclooxygenase (COX)-generated and lipoxygenase (LOX)-generated metabolites have been studied using rodent islets and insulin-secreting cell lines, but very little is known about COX and LOX isoform expression and the effects of modulation of arachidonic acid generation and metabolism in human islets. We have used RT-PCR to identify mRNAs for cytosolic phospholipase A(2) (cPLA(2)), COX-1, COX-2, 5-LOX, and 12-LOX in isolated human islets. COX-3 and 15-LOX were not expressed by human islets. Perifusion experiments with human islets indicated that PLA(2) inhibition inhibited glucose-stimulated insulin secretion, whereas inhibitors of COX-2 and 12-LOX enzymes enhanced basal insulin secretion and also secretory responses induced by 20 mmol/l glucose or by 50 mumol/l arachidonic acid. Inhibition of COX-1 with 100 mumol/l acetaminophen did not significantly affect glucose-stimulated insulin secretion. These data indicate that the stimulation of insulin secretion from human islets in response to arachidonic acid does not require its metabolism through COX-2 and 5-/12-LOX pathways. The products of COX-2 and LOX activities have been implicated in cytokine-mediated damage of beta-cells, so selective inhibitors of these enzymes would be expected to have a dual protective role in diabetes: they would minimize beta-cell dysfunction while maintaining insulin secretion through enhancing endogenous arachidonic acid levels.


Asunto(s)
Ácido Araquidónico/fisiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ácido Araquidónico/metabolismo , Secuencia de Bases , Cadáver , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Cartilla de ADN , Humanos , Secreción de Insulina , Islotes Pancreáticos/enzimología , Lipooxigenasa/genética , Fosfolipasas A/genética , ARN Mensajero/genética , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Pineal Res ; 44(3): 273-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18194202

RESUMEN

Melatonin is known to inhibit insulin secretion from rodent beta-cells through interactions with cell-surface MT1 and/or MT2 receptors, but the function of this hormone in human islets of Langerhans is not known. In the current study, melatonin receptor expression by human islets was examined by reverse transcription-polymerase chain reaction (RT-PCR) and the effects of exogenous melatonin on intracellular calcium ([Ca2+]i) levels and islet hormone secretion were determined by single cell microfluorimetry and radioimmunoassay, respectively. RT-PCR amplifications indicated that human islets express mRNAs coding for MT1 and MT2 melatonin receptors, although MT2 mRNA expression was very low. Analysis of MT1 receptor mRNA expression at the single cell level indicated that it was expressed by human islet alpha-cells, but not by beta-cells. Exogenous melatonin stimulated increases in intracellular calcium ([Ca2+]i) in dissociated human islet cells, and stimulated glucagon secretion from perifused human islets. It also stimulated insulin secretion and this was most probably a consequence of glucagon acting in a paracrine fashion to stimulate beta-cells as the MT1 receptor was absent in beta-cells. Melatonin did not decrease 3', 5'-cyclic adenosine monophosphate (cyclic AMP) levels in human islets, but it inhibited cyclic AMP in the mouse insulinoma (MIN6) beta-cell line and it also inhibited glucose-stimulated insulin secretion from MIN6 cells. These data suggest that melatonin has direct stimulatory effects at human islet alpha-cells and that it stimulates insulin secretion as a consequence of elevated glucagon release. This study also indicates that the effects of melatonin are species-specific with primarily an inhibitory role in rodent beta-cells and a stimulatory effect in human islets.


Asunto(s)
Islotes Pancreáticos/fisiología , Receptores de Melatonina/fisiología , Animales , Línea Celular Tumoral , Colforsina/farmacología , AMP Cíclico/metabolismo , Glucagón/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Ratones , ARN Mensajero/metabolismo , Receptor de Melatonina MT1/biosíntesis , Receptor de Melatonina MT2/biosíntesis , Receptores de Melatonina/biosíntesis , Sistemas de Mensajero Secundario/fisiología
20.
JCI Insight ; 3(3)2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29415896

RESUMEN

Type 1 diabetes (T1D) is a chronic disease characterized by an autoimmune-mediated destruction of insulin-producing pancreatic ß cells. Environmental factors such as viruses play an important role in the onset of T1D and interact with predisposing genes. Recent data suggest that viral infection of human islets leads to a decrease in insulin production rather than ß cell death, suggesting loss of ß cell identity. We undertook this study to examine whether viral infection could induce human ß cell dedifferentiation. Using the functional human ß cell line EndoC-ßH1, we demonstrate that polyinosinic-polycytidylic acid (PolyI:C), a synthetic double-stranded RNA that mimics a byproduct of viral replication, induces a decrease in ß cell-specific gene expression. In parallel with this loss, the expression of progenitor-like genes such as SOX9 was activated following PolyI:C treatment or enteroviral infection. SOX9 was induced by the NF-κB pathway and also in a paracrine non-cell-autonomous fashion through the secretion of IFN-α. Lastly, we identified SOX9 targets in human ß cells as potentially new markers of dedifferentiation in T1D. These findings reveal that inflammatory signaling has clear implications in human ß cell dedifferentiation.


Asunto(s)
Desdiferenciación Celular/inmunología , Diabetes Mellitus Tipo 1/inmunología , Infecciones por Enterovirus/inmunología , Células Secretoras de Insulina/fisiología , Desdiferenciación Celular/efectos de los fármacos , Línea Celular , Diabetes Mellitus Tipo 1/virología , Enterovirus/inmunología , Infecciones por Enterovirus/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Inductores de Interferón/farmacología , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , FN-kappa B/metabolismo , Poli I-C/farmacología , Cultivo Primario de Células , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA