Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(6): 677-686, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31110312

RESUMEN

Consumption of a high-energy Western diet triggers mild adaptive ß cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of ß cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of ß cells, but not that of α cells, leading to enlarged ß cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of ß cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse ß cell failure in patients with diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Animales , Proliferación Celular , Ciclina D2/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Humanos , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Obesidad/tratamiento farmacológico , Parabiosis , Unión Proteica , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
2.
J Biol Chem ; 299(8): 104986, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37392854

RESUMEN

Congenital hyperinsulinism of infancy (CHI) can be caused by a deficiency of the ubiquitously expressed enzyme short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD). To test the hypothesis that SCHAD-CHI arises from a specific defect in pancreatic ß-cells, we created genetically engineered ß-cell-specific (ß-SKO) or hepatocyte-specific (L-SKO) SCHAD knockout mice. While L-SKO mice were normoglycemic, plasma glucose in ß-SKO animals was significantly reduced in the random-fed state, after overnight fasting, and following refeeding. The hypoglycemic phenotype was exacerbated when the mice were fed a diet enriched in leucine, glutamine, and alanine. Intraperitoneal injection of these three amino acids led to a rapid elevation in insulin levels in ß-SKO mice compared to controls. Consistently, treating isolated ß-SKO islets with the amino acid mixture potently enhanced insulin secretion compared to controls in a low-glucose environment. RNA sequencing of ß-SKO islets revealed reduced transcription of ß-cell identity genes and upregulation of genes involved in oxidative phosphorylation, protein metabolism, and Ca2+ handling. The ß-SKO mouse offers a useful model to interrogate the intra-islet heterogeneity of amino acid sensing given the very variable expression levels of SCHAD within different hormonal cells, with high levels in ß- and δ-cells and virtually absent α-cell expression. We conclude that the lack of SCHAD protein in ß-cells results in a hypoglycemic phenotype characterized by increased sensitivity to amino acid-stimulated insulin secretion and loss of ß-cell identity.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasa , Aminoácidos , Hiperinsulinismo Congénito , Hipoglucemia , Secreción de Insulina , Células Secretoras de Insulina , Animales , Ratones , Aminoácidos/metabolismo , Aminoácidos/farmacología , Hipoglucemia/enzimología , Hipoglucemia/genética , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Ratones Noqueados , 3-Hidroxiacil-CoA Deshidrogenasa/deficiencia , 3-Hidroxiacil-CoA Deshidrogenasa/genética , Células Secretoras de Insulina/enzimología , Hiperinsulinismo Congénito/genética
4.
J Biol Chem ; 296: 100646, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33839150

RESUMEN

Dysregulated glucagon secretion deteriorates glycemic control in type 1 and type 2 diabetes. Although insulin is known to regulate glucagon secretion via its cognate receptor (insulin receptor, INSR) in pancreatic alpha cells, the role of downstream proteins and signaling pathways underlying insulin's activities are not fully defined. Using in vivo (knockout) and in vitro (knockdown) studies targeting insulin receptor substrate (IRS) proteins, we compared the relative roles of IRS1 and IRS2 in regulating alpha cell function. Alpha cell-specific IRS1-knockout mice exhibited glucose intolerance and inappropriate glucagon suppression during glucose tolerance tests. In contrast, alpha cell-specific IRS2-knockout animals manifested normal glucose tolerance and suppression of glucagon secretion after glucose administration. Alpha cell lines with stable IRS1 knockdown could not repress glucagon mRNA expression and exhibited a reduction in phosphorylation of AKT Ser/Thr kinase (AKT, at Ser-473 and Thr-308). AlphaIRS1KD cells also displayed suppressed global protein translation, including reduced glucagon expression, impaired cytoplasmic Ca2+ response, and mitochondrial dysfunction. This was supported by the identification of novel IRS1-specific downstream target genes, Trpc3 and Cartpt, that are associated with glucagon regulation in alpha cells. These results provide evidence that IRS1, rather than IRS2, is a dominant regulator of pancreatic alpha cell function.


Asunto(s)
Células Secretoras de Glucagón/patología , Glucagón/metabolismo , Intolerancia a la Glucosa/patología , Proteínas Sustrato del Receptor de Insulina/fisiología , Resistencia a la Insulina , Animales , Femenino , Células Secretoras de Glucagón/metabolismo , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Transducción de Señal
5.
J Biol Chem ; 296: 100495, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33667549

RESUMEN

Human embryonic stem cells are a type of pluripotent stem cells (hPSCs) that are used to investigate their differentiation into diverse mature cell types for molecular studies. The mechanisms underlying insulin receptor (IR)-mediated signaling in the maintenance of human pluripotent stem cell (hPSC) identity and cell fate specification are not fully understood. Here, we used two independent shRNAs to stably knock down IRs in two hPSC lines that represent pluripotent stem cells and explored the consequences on expression of key proteins in pathways linked to proliferation and differentiation. We consistently observed lowered pAKT in contrast to increased pERK1/2 and a concordant elevation in pluripotency gene expression. ERK2 chromatin immunoprecipitation, luciferase assays, and ERK1/2 inhibitors established direct causality between ERK1/2 and OCT4 expression. Of importance, RNA sequencing analyses indicated a dysregulation of genes involved in cell differentiation and organismal development. Mass spectrometry-based proteomic analyses further confirmed a global downregulation of extracellular matrix proteins. Subsequent differentiation toward the neural lineage reflected alterations in SOX1+PAX6+ neuroectoderm and FOXG1+ cortical neuron marker expression and protein localization. Collectively, our data underscore the role of IR-mediated signaling in maintaining pluripotency, the extracellular matrix necessary for the stem cell niche, and regulating cell fate specification including the neural lineage.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Neuronas/citología , Células Madre Pluripotentes/citología , Receptor de Insulina/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Células Madre Embrionarias Humanas/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , Células Madre Pluripotentes/metabolismo , Proteómica/métodos , Transducción de Señal
6.
EMBO J ; 37(24)2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30446598

RESUMEN

A finely tuned balance of self-renewal, differentiation, proliferation, and survival governs the pool size and regenerative capacity of blood-forming hematopoietic stem and progenitor cells (HSPCs). Here, we report that protein kinase C delta (PKCδ) is a critical regulator of adult HSPC number and function that couples the proliferative and metabolic activities of HSPCs. PKCδ-deficient mice showed a pronounced increase in HSPC numbers, increased competence in reconstituting lethally irradiated recipients, enhanced long-term competitive advantage in serial transplantation studies, and an augmented HSPC recovery during stress. PKCδ-deficient HSPCs also showed accelerated proliferation and reduced apoptosis, but did not exhaust in serial transplant assays or induce leukemia. Using inducible knockout and transplantation models, we further found that PKCδ acts in a hematopoietic cell-intrinsic manner to restrict HSPC number and bone marrow regenerative function. Mechanistically, PKCδ regulates HSPC energy metabolism and coordinately governs multiple regulators within signaling pathways implicated in HSPC homeostasis. Together, these data identify PKCδ as a critical regulator of HSPC signaling and metabolism that acts to limit HSPC expansion in response to physiological and regenerative demands.


Asunto(s)
Apoptosis , Médula Ósea/enzimología , Proliferación Celular , Células Madre Hematopoyéticas/enzimología , Proteína Quinasa C-delta/metabolismo , Transducción de Señal , Animales , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Proteína Quinasa C-delta/genética
8.
J Am Soc Nephrol ; 32(9): 2331-2351, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34140396

RESUMEN

BACKGROUND: Mechanisms underlying the pro gression of diabetic kidney disease to ESKD are not fully understood. METHODS: We performed global microRNA (miRNA) analysis on plasma from two cohorts consisting of 375 individuals with type 1 and type 2 diabetes with late diabetic kidney disease, and targeted proteomics analysis on plasma from four cohorts consisting of 746 individuals with late and early diabetic kidney disease. We examined structural lesions in kidney biopsy specimens from the 105 individuals with early diabetic kidney disease. Human umbilical vein endothelial cells were used to assess the effects of miRNA mimics or inhibitors on regulation of candidate proteins. RESULTS: In the late diabetic kidney disease cohorts, we identified 17 circulating miRNAs, represented by four exemplars (miR-1287-5p, miR-197-5p, miR-339-5p, and miR-328-3p), that were strongly associated with 10-year risk of ESKD. These miRNAs targeted proteins in the axon guidance pathway. Circulating levels of six of these proteins-most notably, EFNA4 and EPHA2-were strongly associated with 10-year risk of ESKD in all cohorts. Furthermore, circulating levels of these proteins correlated with severity of structural lesions in kidney biopsy specimens. In contrast, expression levels of genes encoding these proteins had no apparent effects on the lesions. In in vitro experiments, mimics of miR-1287-5p and miR-197-5p and inhibitors of miR-339-5p and miR-328-3p upregulated concentrations of EPHA2 in either cell lysate, supernatant, or both. CONCLUSIONS: This study reveals novel mechanisms involved in progression to ESKD and points to the importance of systemic factors in the development of diabetic kidney disease. Some circulating miRNAs and axon guidance pathway proteins represent potential targets for new therapies to prevent and treat this condition.


Asunto(s)
Orientación del Axón/fisiología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 2/sangre , Nefropatías Diabéticas/etiología , Fallo Renal Crónico/etiología , MicroARNs/sangre , Adulto , Estudios de Cohortes , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/sangre , Femenino , Humanos , Fallo Renal Crónico/sangre , Masculino , Persona de Mediana Edad
9.
Stem Cells ; 38(4): 542-555, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31828876

RESUMEN

A comprehensive characterization of the molecular processes controlling cell fate decisions is essential to derive stable progenitors and terminally differentiated cells that are functional from human pluripotent stem cells (hPSCs). Here, we report the use of quantitative proteomics to describe early proteome adaptations during hPSC differentiation toward pancreatic progenitors. We report that the use of unbiased quantitative proteomics allows the simultaneous profiling of numerous proteins at multiple time points, and is a valuable tool to guide the discovery of signaling events and molecular signatures underlying cellular differentiation. We also monitored the activity level of pathways whose roles are pivotal in the early pancreas differentiation, including the Hippo signaling pathway. The quantitative proteomics data set provides insights into the dynamics of the global proteome during the transition of hPSCs from a pluripotent state toward pancreatic differentiation.


Asunto(s)
Páncreas/metabolismo , Células Madre Pluripotentes/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Diferenciación Celular , Humanos , Páncreas/citología
10.
Diabetologia ; 63(3): 577-587, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31897526

RESUMEN

AIMS/HYPOTHESIS: Sodium-glucose cotransporter 2 (SGLT2) inhibitors, which prevent the renal reabsorption of glucose, decrease blood glucose levels in an insulin-independent manner. We previously reported creating a mouse model of systemic inhibition of target receptors for both insulin and IGF-1 by treating animals with OSI-906, a dual insulin/IGF-1 receptor inhibitor, for 7 days. The OSI-906-treated mice exhibited an increased beta cell mass, hepatic steatosis and adipose tissue atrophy, accompanied by hyperglycaemia and hyperinsulinaemia. In the present study, we investigated the effects of an SGLT2 inhibitor, luseogliflozin, on these changes in OSI-906-treated mice. METHODS: We treated C57BL/6J male mice either with vehicle, luseogliflozin, OSI-906 or OSI-906 plus luseogliflozin for 7 days, and phenotyping was performed to determine beta cell mass and proliferation. Subsequently, we tested whether serum-derived factors have an effect on beta cell proliferation in genetically engineered beta cells, mouse islets or human islets. RESULTS: SGLT2 inhibition with luseogliflozin significantly ameliorated hyperglycaemia, but not hyperinsulinaemia, in the OSI-906-treated mice. Liver steatosis and adipose tissue atrophy induced by OSI-906 were not altered by treatment with luseogliflozin. Beta cell mass and proliferation were further increased by SGLT2 inhibition with luseogliflozin in the OSI-906-treated mice. Luseogliflozin upregulated gene expression related to the forkhead box M1 (FoxM1)/polo-like kinase 1 (PLK1)/centromere protein A (CENP-A) pathway in the islets of OSI-906-treated mice. The increase in beta cell proliferation was recapitulated in a co-culture of Irs2 knockout and Insr/IR knockout (ßIRKO) beta cells with serum from both luseogliflozin- and OSI-906-treated mice, but not after SGLT2 inhibition in beta cells. Circulating factors in both luseogliflozin- and OSI-906-treated mice promoted beta cell proliferation in both mouse islets and cadaveric human islets. CONCLUSIONS/INTERPRETATION: These results suggest that luseogliflozin can increase beta cell proliferation through the activation of the FoxM1/PLK1/CENP-A pathway via humoral factors that act in an insulin/IGF-1 receptor-independent manner.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Sorbitol/análogos & derivados , Animales , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Sinergismo Farmacológico , Técnicas de Inactivación de Genes , Humanos , Imidazoles/farmacología , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Pirazinas/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sorbitol/farmacología
11.
J Biol Chem ; 294(3): 1059-1069, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30459233

RESUMEN

FoxO proteins are major targets of insulin action, and FoxO1 mediates the effects of insulin on hepatic glucose metabolism. We reported previously that serpinB1 is a liver-secreted factor (hepatokine) that promotes adaptive ß-cell proliferation in response to insulin resistance in the liver-specific insulin receptor knockout (LIRKO) mouse. Here we report that FoxO1 plays a critical role in promoting serpinB1 expression in hepatic insulin resistance in a non-cell-autonomous manner. Mice lacking both the insulin receptor and FoxO1 (LIRFKO) exhibit reduced ß-cell mass compared with LIRKO mice because of attenuation of ß-cell proliferation. Although hepatic expression of serpinB1 mRNA and protein levels was increased in LIRKO mice, both the mRNA and protein levels returned to control levels in LIRFKO mice. Furthermore, liver-specific expression of constitutively active FoxO1 in transgenic mice induced an increase in hepatic serpinB1 mRNA and protein levels in refed mice. Conversely, serpinB1 mRNA and protein levels were reduced in mice lacking FoxO proteins in the liver. ChIP studies demonstrated that FoxO1 binds to three distinct sites located ∼9 kb upstream of the serpinb1 gene in primary mouse hepatocytes and that this binding is enhanced in hepatocytes from LIRKO mice. However, adenoviral expression of WT or constitutively active FoxO1 and insulin treatment are sufficient to regulate other FoxO1 target genes (IGFBP-1 and PEPCK) but not serpinB1 expression in mouse primary hepatocytes. These results indicate that liver FoxO1 promotes serpinB1 expression in hepatic insulin resistance and that non-cell-autonomous factors contribute to FoxO1-dependent effects on serpinB1 expression in the liver.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hígado/metabolismo , Serpinas/biosíntesis , Animales , Proteína Forkhead Box O1/genética , Hepatocitos/citología , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Hígado/citología , Masculino , Ratones , Ratones Transgénicos , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Serpinas/genética
12.
J Am Chem Soc ; 142(14): 6477-6482, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32175731

RESUMEN

The loss of insulin-producing ß-cells is the central pathological event in type 1 and 2 diabetes, which has led to efforts to identify molecules to promote ß-cell proliferation, protection, and imaging. However, the lack of ß-cell specificity of these molecules jeopardizes their therapeutic potential. A general platform for selective release of small-molecule cargoes in ß-cells over other islet cells ex vivo or other cell-types in an organismal context will be immensely valuable in advancing diabetes research and therapeutic development. Here, we leverage the unusually high Zn(II) concentration in ß-cells to develop a Zn(II)-based prodrug system to selectively and tracelessly deliver bioactive small molecules and fluorophores to ß-cells. The Zn(II)-targeting mechanism enriches the inactive cargo in ß-cells as compared to other pancreatic cells; importantly, Zn(II)-mediated hydrolysis triggers cargo activation. This prodrug system, with modular components that allow for fine-tuning selectivity, should enable the safer and more effective targeting of ß-cells.


Asunto(s)
Linfocitos B/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Zinc/uso terapéutico , Catálisis , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Humanos
13.
Mol Cell ; 45(6): 826-35, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22387028

RESUMEN

Although expression of the mammalian RNA-binding protein HuD was considered to be restricted to neurons, we report that HuD is present in pancreatic ß cells, where its levels are controlled by the insulin receptor pathway. We found that HuD associated with a 22-nucleotide segment of the 5' untranslated region (UTR) of preproinsulin (Ins2) mRNA. Modulating HuD abundance did not alter Ins2 mRNA levels, but HuD overexpression decreased Ins2 mRNA translation and insulin production, and conversely, HuD silencing enhanced Ins2 mRNA translation and insulin production. Following treatment with glucose, HuD rapidly dissociated from Ins2 mRNA and enabled insulin biosynthesis. Importantly, HuD-knockout mice displayed higher insulin levels in pancreatic islets, while HuD-overexpressing mice exhibited lower insulin levels in islets and in plasma. In sum, our results identify HuD as a pivotal regulator of insulin translation in pancreatic ß cells.


Asunto(s)
Proteínas ELAV/metabolismo , Insulina/genética , Insulina/metabolismo , Biosíntesis de Proteínas , Regiones no Traducidas 5' , Animales , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Glucosa/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Precursores de Proteínas/genética
15.
J Biol Chem ; 293(16): 5934-5946, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29496993

RESUMEN

Chronic low-grade inflammation in the pancreatic islets is observed in individuals with type 2 diabetes, and macrophage levels are elevated in the islets of these individuals. However, the molecular mechanisms underlying the interactions between the pancreatic ß cells and macrophages and their involvement in inflammation are not fully understood. Here, we investigated the role of S100 calcium-binding protein A8 (S100A8), a member of the damage-associated molecular pattern molecules (DAMPs), in ß-cell inflammation. Co-cultivation of pancreatic islets with unstimulated peritoneal macrophages in the presence of palmitate (to induce lipotoxicity) and high glucose (to induce glucotoxicity) synergistically increased the expression and release of islet-produced S100A8 in a Toll-like receptor 4 (TLR4)-independent manner. Consistently, a significant increase in the expression of the S100a8 gene was observed in the islets of diabetic db/db mice. Furthermore, the islet-derived S100A8 induced TLR4-mediated inflammatory cytokine production by migrating macrophages. When human islet cells were co-cultured with U937 human monocyte cells, the palmitate treatment up-regulated S100A8 expression. This S100A8-mediated interaction between islets and macrophages evoked ß-cell apoptosis, which was ameliorated by TLR4 inhibition in the macrophages or S100A8 neutralization in the pancreatic islets. Of note, both glucotoxicity and lipotoxicity triggered S100A8 secretion from the pancreatic islets, which in turn promoted macrophage infiltration of the islets. Taken together, a positive feedback loop between islet-derived S100A8 and macrophages drives ß-cell apoptosis and pancreatic islet inflammation. We conclude that developing therapeutic approaches to inhibit S100A8 may serve to prevent ß-cell loss in patients with diabetes.


Asunto(s)
Apoptosis , Calgranulina A/inmunología , Inflamación/inmunología , Células Secretoras de Insulina/inmunología , Macrófagos/inmunología , Animales , Línea Celular , Células Cultivadas , Glucosa/inmunología , Humanos , Células Secretoras de Insulina/citología , Macrófagos/citología , Masculino , Ratones Endogámicos C57BL , Palmitatos/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología
16.
Anal Chem ; 91(9): 5794-5801, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843680

RESUMEN

Comprehensive phosphoproteomic analysis of small populations of cells remains a daunting task due primarily to the insufficient MS signal intensity from low concentrations of enriched phosphopeptides. Isobaric labeling has a unique multiplexing feature where the "total" peptide signal from all channels (or samples) triggers MS/MS fragmentation for peptide identification, while the reporter ions provide quantitative information. In light of this feature, we tested the concept of using a "boosting" sample (e.g., a biological sample mimicking the study samples but available in a much larger quantity) in multiplexed analysis to enable sensitive and comprehensive quantitative phosphoproteomic measurements with <100 000 cells. This simple boosting to amplify signal with isobaric labeling (BASIL) strategy increased the overall number of quantifiable phosphorylation sites more than 4-fold. Good reproducibility in quantification was demonstrated with a median CV of 15.3% and Pearson correlation coefficient of 0.95 from biological replicates. A proof-of-concept experiment demonstrated the ability of BASIL to distinguish acute myeloid leukemia cells based on the phosphoproteome data. Moreover, in a pilot application, this strategy enabled quantitative analysis of over 20 000 phosphorylation sites from human pancreatic islets treated with interleukin-1ß and interferon-γ. Together, this signal boosting strategy provides an attractive solution for comprehensive and quantitative phosphoproteome profiling of relatively small populations of cells where traditional phosphoproteomic workflows lack sufficient sensitivity.


Asunto(s)
Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/metabolismo , Fosfopéptidos/metabolismo , Fosfoproteínas/metabolismo , Coloración y Etiquetado/métodos , Espectrometría de Masas en Tándem/métodos , Antivirales/farmacología , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Fosforilación
17.
Curr Diab Rep ; 19(8): 48, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31250214

RESUMEN

PURPOSE OF REVIEW: Pancreatic ß-cells play a critical role in whole-body glucose homeostasis by regulating the release of insulin in response to minute by minute alterations in metabolic demand. As such, ß-cells are staunchly resilient but there are circumstances where they can become functionally compromised or physically lost due to pathophysiological changes which culminate in overt hyperglycemia and diabetes. RECENT FINDINGS: In humans, ß-cell mass appears to be largely defined in the postnatal period and this early replicative and generative phase is followed by a refractory state which persists throughout life. Despite this, efforts to identify physiological and pharmacological factors which might re-initiate ß-cell replication (or cause the replenishment of ß-cells by neogenesis or transdifferentiation) are beginning to bear fruit. Controlled manipulation of ß-cell mass in humans still represents a holy grail for therapeutic intervention in diabetes, but progress is being made which may lead to ultimate success.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Células Secretoras de Insulina , Humanos , Insulina
18.
J Pathol ; 246(2): 231-243, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30014466

RESUMEN

For the majority of patients diagnosed with pancreatic neuroendocrine tumors (NETs), there is significant malignant potential with a poor prognosis; however, the molecular abnormalities and pathogenesis of pancreatic NETs have not been firmly established. Here, we report that loss of expression of the RNA-binding protein HuD correlates with low p27Kip1 (p27) levels and poor prognosis in pancreatic NETs. HuD expression was frequently lost in many human pancreatic NETs, and these pancreatic NETs showed aggressive clinicopathological phenotypes with low p27 levels, increased tumor size, higher World Health Organization grade and pT stage of the tumor, and the presence of angioinvasion. Furthermore, loss of HuD was an independent, progression-free prognostic factor in multivariate survival analysis. However, the level of HuR, a member of the same Hu protein family as HuD, was not significantly correlated with pancreatic NET size and progression. Mechanistically, HuD enhanced p27 mRNA translation by interacting with both the 5'-untranslated region (UTR) and the 3'-UTR of p27 mRNA, and consequently suppressed cell cycle progression and tumor growth. In addition, HuD competed with miR-30a-3p for binding to the 3'-UTR of p27 mRNA, suggesting an interplay between HuD and miR-30a-3p in controlling p27 translation. Our results identify HuD as a pivotal suppressor of pancreatic NET growth, and suggest that HuD has potential value as a prognostic factor of pancreatic NETs. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Neuroendocrino/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína 4 Similar a ELAV/metabolismo , Neoplasias Pancreáticas/metabolismo , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Adulto , Anciano , Animales , Sitios de Unión , Biomarcadores de Tumor/genética , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/mortalidad , Carcinoma Neuroendocrino/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo , Proteína 4 Similar a ELAV/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Fenotipo , Supervivencia sin Progresión , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal , Factores de Tiempo , Carga Tumoral
19.
J Cell Mol Med ; 22(4): 2337-2345, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29431265

RESUMEN

Cannabinoid 1 receptors (CB1Rs) are expressed in peripheral tissues, including islets of Langerhans, where their function(s) is under scrutiny. Using mouse ß-cell lines, human islets and CB1R-null (CB1R-/- ) mice, we have now investigated the role of CB1Rs in modulating ß-cell function and glucose responsiveness. Synthetic CB1R agonists diminished GLP-1-mediated cAMP accumulation and insulin secretion as well as glucose-stimulated insulin secretion in mouse ß-cell lines and human islets. In addition, silencing CB1R in mouse ß cells resulted in an increased expression of pro-insulin, glucokinase (GCK) and glucose transporter 2 (GLUT2), but this increase was lost in ß cells lacking insulin receptor. Furthermore, CB1R-/- mice had increased pro-insulin, GCK and GLUT2 expression in ß cells. Our results suggest that CB1R signalling in pancreatic islets may be harnessed to improve ß-cell glucose responsiveness and preserve their function. Thus, our findings further support that blocking peripheral CB1Rs would be beneficial to ß-cell function in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor Cannabinoide CB1/genética , Animales , Antígenos CD/genética , AMP Cíclico/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica/genética , Glucoquinasa/genética , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/genética , Humanos , Insulina/genética , Células Secretoras de Insulina/patología , Ratones , Receptor Cannabinoide CB1/metabolismo , Receptor de Insulina/genética
20.
J Biol Chem ; 292(43): 17598-17608, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28860191

RESUMEN

Preservation of insulin-secreting ß-cells is an important goal for therapies aimed at restoring normoglycemia in patients with diabetes. One approach, the inhibition of histone deacetylases (HDACs), has been reported to suppress pancreatic islet inflammation and ß-cell apoptosis in vitro In this report, we demonstrate the efficacy of HDAC inhibitors (HDACi) in vivo We show that daily administration of BRD3308, an isoform-selective HDAC3 inhibitor, for 2 weeks to female nonobese diabetic (NOD) mice, beginning at 3 weeks of age, followed by twice-weekly injections until age 25 weeks, protects the animals from diabetes. The preservation of ß-cells was because of a significant decrease in islet infiltration of mononuclear cells. Moreover, the BRD3308 treatment increased basal insulin secretion from islets cultured in vitro All metabolic tissues tested in vehicle- or BRD3308-treated groups showed virtually no sign of immune cell infiltration, except minimal infiltration in white adipose tissue in animals treated with the highest BRD3308 dose (10 mg/kg), providing additional evidence of protection from immune attack in the treated groups. Furthermore, pancreata from animals treated with 10 mg/kg BRD3308 exhibited significantly decreased numbers of apoptotic ß-cells compared with those treated with vehicle or low-dose BRD3308. Finally, animals treated with 1 or 10 mg/kg BRD3308 had enhanced ß-cell proliferation. These in vivo results point to the potential use of selective HDAC3 inhibitors as a therapeutic approach to suppress pancreatic islet infiltration and prevent ß-cell death with the long-term goal of limiting the progression of type 1 diabetes.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/prevención & control , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Células Secretoras de Insulina/enzimología , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/patología , Animales , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Femenino , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA