Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Intervalo de año de publicación
1.
Inflamm Regen ; 42(1): 52, 2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36447218

RESUMEN

BACKGROUND: Different factors may lead to hepatitis. Among which are liver inflammation and poisoning. We chose two hepatitis models, typical for these two underlying causes. Thus, we aimed to characterize the role of protease-activated receptor 2 (Par2) in liver regeneration and inflammation to reconcile Par2 conflicting role in many damage models, which sometimes aggravates the induced damage and sometimes alleviates it. METHODS: WT and knockout (Par2KO) mice were injected with concanavalin A (ConA) to induce immune-mediated hepatitis or with carbon tetrachloride (CCl4) to elicit direct hepatic damage. To distinguish the immune component from the liver regenerative response, we conducted bone marrow (BM) replacements of WT and Par2KO mice and repeated the damage models. RESULTS: ConA injection caused limited damage in Par2KO mice livers, while in the WT mice severe damage followed by leukocyte infiltration was evident. Reciprocal BM replacement of WT and Par2KO showed that WT BM-reconstituted Par2KO mice displayed marked liver damage, while in Par2KO BM-reconstituted WT mice, the tissue was generally protected. In the CCl4 direct damage model, hepatocytes regenerated in WT mice, whereas Par2KO mice failed to recover. Reciprocal BM replacement did not show significant differences in hepatic regeneration. In Par2KO mice, hepatitis was more apparent, while WT recovered regardless of the BM origin. CONCLUSIONS: We conclude that Par2 activation in the immune system aggravates hepatitis and that Par2 activation in the damaged tissue promotes liver regeneration. When we incorporate this finding and revisit the literature reports, we reconciled the conflicts surrounding Par2's role in injury, recovery, and inflammation.

2.
PLoS One ; 17(4): e0266066, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35385524

RESUMEN

HNF4α has been implicated in IBD through a number of genome-wide association studies. Recently, we developed potent HNF4α agonists, including N-trans caffeoyltyramine (NCT). NCT was identified by structural similarity to previously the previously identified but weak HNF4α agonists alverine and benfluorex. Here, we administered NCT to mice fed a high fat diet, with the goal of studying the role of HNF4α in obesity-related diseases. Intestines from NCT-treated mice were examined by RNA-seq to determine the role of HNF4α in that organ. Surprisingly, the major classes of genes altered by HNF4α were involved in IBD and Paneth cell biology. Multiple genes downregulated in IBD were induced by NCT. Paneth cells identified by lysozyme expression were reduced in high fat fed mice. NCT reversed the effect of high fat diet on Paneth cells, with multiple markers being induced, including a number of defensins, which are critical for Paneth cell function and intestinal barrier integrity. NCT upregulated genes that play important role in IBD and that are downregulated in that disease. It reversed the loss of Paneth cell markers that occurred in high fat diet fed mice. These data suggest that HNF4α could be a therapeutic target for IBD and that the agonists that we have identified could be candidate therapeutics.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Células de Paneth , Animales , Estudio de Asociación del Genoma Completo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Ratones , Células de Paneth/metabolismo
3.
Biomedicines ; 10(3)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35327373

RESUMEN

ß-cell number and/or function is reduced in diabetes. Thus, inducing the formation of new ß-cells has been a major goal of diabetes research. However, the pathway(s) by which new ß-cells form when preexisting ß-cells are decreased in number or cease to function has remained obscure. Many pathways have been proposed, but definitive evidence, particularly in humans, has been lacking. Replication of preexisting ß-cells, neogenesis from ducts, redifferentiation from ß-cells that dedifferentiated under metabolic stress, and transdifferentiation from other cell types, particularly within the islet, are the major mechanisms that have been proposed for generating increased numbers of functional ß-cells. Here, I will discuss those approaches critically, with particular attention to transdifferentiation of preexisting α-cells to ß-cells.

4.
Cell Death Dis ; 13(1): 89, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35087037

RESUMEN

We report here that the potent HNF4α agonist N-trans-caffeoyltyramine (NCT) promotes weight loss by inducing an increase in mitochondrial mass and function, including fatty acid oxidation. Previously, we found in a short term trial in obese mice that NCT promoted reversal of hepatic steatosis through a mechanism involving the stimulation of lipophagy by dihydroceramides. NCT led to increased dihydroceramide levels by inhibiting dihydroceramide conversion to ceramides. Here, we were able to administer NCT orally, permitting longer term administration. Mice fed NCT mixed with high fat diet exhibited decreased weight. Examination of RNA-seq data revealed an increase in PPARGC1A, a central regulator of mitochondrial biogenesis. In addition to the decreased hepatic steatosis that we found previously, mice fed a high fat diet containing NCT mice weighed substantially less than control mice fed high fat diet alone. They had increased mitochondrial mass, exhibited increased fatty acid oxidation, and had an increased level of NAD. Markers of liver inflammation such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα), which are important in the progression of non-alcoholic fatty liver disease to non-alcoholic steatohepatitis were decreased by NCT. There was no evidence of any toxicity from NCT consumption. These results indicate that HNF4α is an important regulator of mitochondrial mass and function and support that use of HNF4α to treat disorders of fatty acid excess, potentially including obesity, NAFLD, and NASH.


Asunto(s)
Hígado , Enfermedad del Hígado Graso no Alcohólico , Administración Oral , Animales , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/patología , Aumento de Peso
5.
Cell Death Dis ; 12(6): 603, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117215

RESUMEN

We report the discovery of strong HNF4α agonists and their use to uncover a previously unknown pathway by which HNF4α controls the level of fat storage in the liver. This involves the induction of lipophagy by dihydroceramides, the synthesis and secretion of which is controlled by genes induced by HNF4α. The HNF4α activators are N-trans caffeoyltyramine (NCT) and N-trans feruloyltyramine (NFT), which are structurally related to the known drugs alverine and benfluorex, which we previously showed to be weak HNF4α activators. In vitro, NCT and NFT induced fat clearance from palmitate-loaded cells. In DIO mice, NCT led to recovery of hepatic HNF4α expression and reduction of steatosis. Mechanistically, increased dihydroceramide production and action downstream of HNF4α occurred through increased expression of HNF4α downstream genes, including SPNS2 and CYP26A1. NCT was completely nontoxic at the highest dose administered and so is a strong candidate for an NAFLD therapeutic.


Asunto(s)
Ácidos Cafeicos/farmacología , Factor Nuclear 4 del Hepatocito/fisiología , Metabolismo de los Lípidos , Hígado/metabolismo , Tiramina/análogos & derivados , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Células Cultivadas , Ácidos Cumáricos/farmacología , Células HeLa , Células Hep G2 , Factor Nuclear 4 del Hepatocito/agonistas , Factor Nuclear 4 del Hepatocito/genética , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Tiramina/farmacología
6.
Islets ; : 1-12, 2018 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-29723131

RESUMEN

Recently, we showed that pancreatitis in the context of profound ß-cell deficiency was sufficient to induce islet cell transdifferentiation. In some circumstances, this effect was sufficient to result in recovery from severe diabetes. More recently, we showed that the molecular mechanism by which pancreatitis induced ß-cell neogenesis by transdifferentiation was activation of an atypical GPCR called Protease-Activated Receptor 2 (PAR2). However, the ability of PAR2 to induce transdifferentiation occurred only in the setting of profound ß-cell deficiency, implying the existence of a repressive factor from those cells. Here we show that the repressor from ß-cells is insulin. Treatment of primary islets with a PAR2 agonist (2fLI) in combination with inhibitors of insulin secretion and signaling was sufficient to induce insulin and PAX4 gene expression. Moreover, in primary human islets, this treatment also led to the induction of bihormonal islet cells coexpressing glucagon and insulin, a hallmark of islet cell transdifferentiation. Mechanistically, insulin inhibited the positive effect of a PAR2 agonist on insulin gene expression and also led to an increase in PAX4, which plays an important role in islet cell transdifferentiation. The studies presented here demonstrate that insulin represses transdifferentiation of α- to ß-cells induced by activation of PAR2. This provides a mechanistic explanation for the observation that α- to ß-cell transdifferentiation occurs only in the setting of severe ß-cell ablation. The mechanistic understanding of islet cell transdifferentiation and the ability to modulate that process using available pharmacological reagents represents an important step along the path towards harnessing this novel mechanism of ß-cell neogenesis as a therapy for diabetes.

7.
Biochem Pharmacol ; 138: 140-149, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28522407

RESUMEN

Pancreatic ß-cell lipotoxicity is a central feature of the pathogenesis of type 2 diabetes. To study the mechanism by which fatty acids cause ß-cell death and develop novel approaches to prevent it, a high-throughput screen on the ß-cell line INS1 was carried out. The cells were exposed to palmitate to induce cell death and compounds that reversed palmitate-induced cytotoxicity were ascertained. Hits from the screen were analyzed by an increasingly more stringent testing funnel, ending with studies on primary human islets treated with palmitate. MAP4K4 inhibitors, which were not part of the screening libraries but were ascertained by a bioinformatics analysis, and the endocannabinoid anandamide were effective at inhibiting palmitate-induced apoptosis in INS1 cells as well as primary rat and human islets. These targets could serve as the starting point for the development of therapeutics for type 2 diabetes.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Línea Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Biología Computacional , Ácidos Grasos no Esterificados/efectos adversos , Ácidos Grasos no Esterificados/antagonistas & inhibidores , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Ratas Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Técnicas de Cultivo de Tejidos
8.
Cell Death Dis ; 7(11): e2452, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27809303

RESUMEN

Understanding the mechanisms by which cells sense and respond to injury is central to developing therapies to enhance tissue regeneration. Previously, we showed that pancreatic injury consisting of acinar cell damage+ß-cell ablation led to islet cell transdifferentiation. Here, we report that the molecular mechanism for this requires activating protease-activated receptor-2 (PAR2), a G-protein-coupled receptor. PAR2 modulation was sufficient to induce islet cell transdifferentiation in the absence of ß-cells. Its expression was modulated in an islet cell type-specific manner in murine and human type 1 diabetes (T1D). In addition to transdifferentiation, PAR2 regulated ß-cell apoptosis in pancreatitis. PAR2's role in regeneration is broad, as mice lacking PAR2 had marked phenotypes in response to injury in the liver and in digit regeneration following amputation. These studies provide a pharmacologically relevant target to induce tissue regeneration in a number of diseases, including T1D.


Asunto(s)
Transdiferenciación Celular , Receptor PAR-2/metabolismo , Regeneración , Animales , Tetracloruro de Carbono , Muerte Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Ceruletida/farmacología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Extremidades , Regulación de la Expresión Génica/efectos de los fármacos , Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción Paired Box/metabolismo , Pancreatitis/metabolismo , Pancreatitis/patología , Regeneración/efectos de los fármacos , Factores de Transcripción/metabolismo
9.
Physiol Rep ; 2(9)2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25194022

RESUMEN

The maternal embryonic leucine zipper kinase (MELK) is expressed in stem/progenitor cells in some adult tissues, where it has been implicated in diverse biological processes, including the control of cell proliferation. Here, we described studies on its role in adult pancreatic regeneration in response to injury induced by duct ligation and ß-cell ablation. MELK expression was studied using transgenic mice expressing GFP under the control of the MELK promoter, and the role of MELK was studied using transgenic mice deleted in the MELK kinase domain. Pancreatic damage was initiated using duct ligation and chemical beta-cell ablation. By tracing MELK expression using a MELK promoter-GFP transgene, we determined that expression was extremely low in the normal pancreas. However, following duct ligation and ß-cell ablation, it became highly expressed in pancreatic ductal cells while remaining weakly expressed in α-cells and ß- cells. In a mutant mouse in which the MELK kinase domain was deleted, there was no effect on pancreatic development. There was no apparent effect on islet regeneration, either. However, following duct ligation there was a dramatic increase in the number of small ducts, but no change in the total number of duct cells or duct cell proliferation. In vitro studies indicated that this was likely due to a defect in cell migration. These results implicate MELK in the control of the response of the pancreas to injury, specifically controlling cell migration in normal and transformed pancreatic duct cells.

10.
Stem Cells ; 31(11): 2388-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922239

RESUMEN

Achieving efficient ß-cell regeneration is a major goal of diabetes research. Previously, we found that a combination of ß-cell ablation and pancreatic duct ligation led to ß-cell regeneration by direct conversion from α-cells. Here, we studied the effect of surgical reversal of the duct ligation, finding that there was a wave of ß-cell replication following reversal. The combination of ß-cell neogenesis prior to reversal of the duct ligation and ß-cell replication following reversal resulted in efficient ß-cell regeneration and eventual recovery of function. This provides an important proof of principle that efficient ß-cell regeneration is possible, even from a starting point of profound ß-cell ablation. This has important implications for efforts to promote ß-cell regeneration.


Asunto(s)
Técnicas de Ablación/métodos , Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus/cirugía , Células Secretoras de Insulina/citología , Islotes Pancreáticos/cirugía , Conductos Pancreáticos/cirugía , Animales , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Stem Cells ; 31(11): 2396-407, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922283

RESUMEN

Increasing the number of ß cells is critical to a definitive therapy for diabetes. Previously, we discovered potent synthetic small molecule antagonists of the nuclear receptor transcription factor HNF4α. The natural ligands of HNF4α are thought to be fatty acids. Because obesity, in which there are high circulating levels of free fatty acids, is one of the few conditions leading to ß-cell hyperplasia, we tested the hypothesis that a potent HNF4α antagonist might stimulate ß-cell replication. A bioavailable HNF4α antagonist was injected into normal mice and rabbits and ß-cell ablated mice and the effect on ß-cell replication was measured. In normal mice and rabbits, the compound induced ß-cell replication and repressed the expression of multiple cyclin-dependent kinase inhibitors, including p16 that plays a critical role in suppressing ß-cell replication. Interestingly, in ß-cell ablated mice, the compound induced α- and δ-cell, in addition to ß-cell replication, and ß-cell number was substantially increased. Overall, the data presented here are consistent with a model in which the well-known effects of obesity and high fat diet on ß-cell replication occur by inhibition of HNF4α. The availability of a potent synthetic HNF4α antagonist raises the possibility that this effect might be a viable route to promote significant increases in ß-cell replication in diseases with reduced ß-cell mass, including type I and type II diabetes.


Asunto(s)
Factor Nuclear 4 del Hepatocito/antagonistas & inhibidores , Células Secretoras de Insulina/metabolismo , Ácido Oléico/farmacología , Ácidos Palmíticos/farmacología , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Células Hep G2 , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Conejos , Distribución Aleatoria
12.
ACS Chem Biol ; 8(8): 1730-6, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23675775

RESUMEN

The principal finding of this study is that two drugs, alverine and benfluorex, used in vastly different clinical settings, activated the nuclear receptor transcription factor HNF4α. Both were hits in a high-throughput screen for compounds that reversed the inhibitory effect of the fatty acid palmitate on human insulin promoter activity. Alverine is used in the treatment of irritable bowel syndrome, while benfluorex (Mediator) was used to treat hyperlipidemia and type II diabetes. Benfluorex was withdrawn from the market recently because of serious cardiovascular side effects related to fenfluramine-like activity. Strikingly, alverine and benfluorex have a previously unrecognized structural similarity, consistent with a common mechanism of action. Gene expression and biochemical studies revealed that they both activate HNF4α. This novel mechanism of action should lead to a reinterpretation of previous studies with these drugs and suggests a path toward the development of therapies for diseases such as inflammatory bowel and diabetes that may respond to HNF4α activators.


Asunto(s)
Fenfluramina/análogos & derivados , Factor Nuclear 4 del Hepatocito/metabolismo , Propilaminas/química , Línea Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Fenfluramina/química , Fenfluramina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Moleculares , Estructura Molecular , Propilaminas/farmacología , Unión Proteica/efectos de los fármacos
13.
Chem Biol ; 19(7): 806-18, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22840769

RESUMEN

Hepatocyte nuclear factor (HNF)4α is a central regulator of gene expression in cell types that play a critical role in metabolic homeostasis, including hepatocytes, enterocytes, and pancreatic ß cells. Although fatty acids were found to occupy the HNF4α ligand-binding pocket and were proposed to act as ligands, there is controversy about both the nature of HNF4α ligands as well as the physiological role of the binding. Here, we report the discovery of potent synthetic HNF4α antagonists through a high-throughput screen for effectors of the human insulin promoter. These molecules bound to HNF4α with high affinity and modulated the expression of known HNF4α target genes. Notably, they were found to be selectively cytotoxic to cancer cell lines in vitro and in vivo, although in vivo potency was limited by suboptimal pharmacokinetic properties. The discovery of bioactive modulators for HNF4α raises the possibility that diseases involving HNF4α, such as diabetes and cancer, might be amenable to pharmacologic intervention by modulation of HNF4α activity.


Asunto(s)
Bencimidazoles/farmacología , Descubrimiento de Drogas , Factor Nuclear 4 del Hepatocito/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Insulina/genética , Regiones Promotoras Genéticas/genética , Sulfonamidas/farmacología , Bencimidazoles/química , Relación Dosis-Respuesta a Droga , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , PPAR gamma/agonistas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Relación Estructura-Actividad , Sulfonamidas/química
14.
Islets ; 3(6): 358-66, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21964314

RESUMEN

Elucidating mechanisms of cell cycle control in normally quiescent human pancreatic ß-cells has the potential to impact regeneration strategies for diabetes. Previously we demonstrated that Id3, a repressor of basic Helix-Loop-Helix (bHLH) proteins, was sufficient to induce cell cycle entry in pancreatic duct cells, which are closely related to ß-cells developmentally. We hypothesized that Id3 might similarly induce cell cycle entry in primary human ß-cells. To test this directly, adult human ß-cells were transduced with adenovirus expressing Id3. Consistent with a replicative response, ß-cells exhibited BrdU incorporation. Further, Id3 potently repressed expression of the cyclin dependent kinase inhibitor p57 (Kip2 ) , a gene which is also silenced in a rare ß-cell hyperproliferative disorder in infants. Surprisingly however, BrdU positive ß-cells did not express the proliferation markers Ki67 and pHH3. Instead, BrdU uptake reflected a DNA damage response, as manifested by hydroxyurea incorporation, γH2AX expression, and 53BP1 subcellular relocalization. The uncoupling of BrdU uptake from replication raises a cautionary note about interpreting studies relying solely upon BrdU incorporation as evidence of ß-cell proliferation. The data also establish that loss of p57 (Kip2) is not sufficient to induce cell cycle entry in adult ß-cells. Moreover, the differential responses to Id3 between duct and ß-cells reveal that ß-cells possess intrinsic resistance to cell cycle entry not common to all quiescent epithelial cells in the adult human pancreas. The data provide a much needed comparative model for investigating the molecular basis for this resistance in order to develop a strategy for improving replication competence in ß-cells.


Asunto(s)
Bromodesoxiuridina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Daño del ADN , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Diferenciación/genética , Células Secretoras de Insulina/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Transfección , Proteína 1 de Unión al Supresor Tumoral P53 , Regulación hacia Arriba
15.
Islets ; 3(6): 327-37, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21975561

RESUMEN

Glucose homeostasis depends on adequate control of insulin secretion. We report the association of the cell-adhesion and adiponectin (APN)-binding glycoprotein T-cadherin (Cdh13) with insulin granules in mouse and human ß-cells. Immunohistochemistry and electron microscopy of islets in situ and targeting of RFP-tagged T-cadherin to GFP-labeled insulin granules in isolated ß-cells demonstrate this unusual location. Analyses of T-cadherin-deficient (Tcad-KO) mice show normal islet architecture and insulin content. However, T-cadherin is required for sufficient insulin release in vitro and in vivo. Primary islets from Tcad-KO mice were defective in glucose-induced but not KCl-mediated insulin secretion. In vivo, second phase insulin release in T-cad-KO mice during a hyperglycemic clamp was impaired while acute first phase release was unaffected. Tcad-KO mice showed progressive glucose intolerance by 5 mo of age without concomitant changes in peripheral insulin sensitivity. Our analyses detected no association of APN with T-cadherin on ß-cell granules although colocalization was observed on the pancreatic vasculature. These data identify T-cadherin as a novel component of insulin granules and suggest that T-cadherin contributes to the regulation of insulin secretion independently of direct interactions with APN.


Asunto(s)
Cadherinas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Adiponectina/metabolismo , Animales , Western Blotting , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Humanos , Inmunohistoquímica , Secreción de Insulina , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Inmunoelectrónica
16.
Islets ; 3(4): 188-95, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21623169

RESUMEN

Partial pancreatectomy is one of the most commonly used models in the study of ß-cell regeneration. The mechanism by which regeneration occurs in this model has been controversial, with some claiming that islet and ß-cell neogenesis is important, while others claim that ß-cell replication is predominant. Here, we combined a time course analysis with continuous BrdU administration to study ß-cell regeneration following partial pancreatectomy. While exocrine cells in regenerating areas were highly proliferative and positive for BrdU, islets in regenerating areas were negative for BrdU one week after partial pancreatectomy, suggesting that they were derived from preexisting islets rather than being neogenic. The insulin-positive cells in ducts that have been reported by others and taken as evidence of ß-cell neogenesis were present in regenerating regions of the pancreas, but were relatively uncommon and were not highly proliferative, suggesting that they could not account for significant islet neogenesis. Consistent with a lack of islet neogenesis, regenerating areas following a second partial pancreatectomy were devoid of islets. ß-cell replication was detectable at a high frequency two weeks following partial pancreatectomy and was present at a similar frequency in both regenerating and preexisting regions of the pancreas. In summary, our data indicate that islet neogenesis following partial pancreatectomy does not occur.


Asunto(s)
Proliferación Celular , Replicación del ADN , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Pancreatectomía , Regeneración , Animales , Bromodesoxiuridina/farmacocinética , Recuento de Células , Diabetes Mellitus/fisiopatología , Diabetes Mellitus/terapia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Inmunohistoquímica , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos ICR , ARN Mensajero , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
17.
Mol Cancer Res ; 9(6): 782-90, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21498546

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) has a 5-year survival rate of less than 5%, and therapeutic advances have been hampered by gaps in our understanding of cell-cycle control in the adult pancreas. Previously, we reported that basic Helix-Loop-Helix (bHLH) transcription factors regulate cell fate specification in the pancreas. In the present study, we found that a repressor of bHLH activity, Id3, was profoundly upregulated in ductal cells in murine models of pancreatitis and pancreatic intraepithelial neoplasia (PanIN). Id3 was also pervasively expressed in neoplastic lesions in human PDA in situ. We hypothesized that an imbalance in bHLH versus Id activity controlled cell growth in PDA. Consistent with this model, cell-cycle progression in PDA cells was impeded by siRNA-mediated depletion of Id3 or overexpression of the bHLH protein E47. The precursors of human PDA are normally quiescent duct cells which do not proliferate in response to high serum or growth factors. The finding that Id3 was expressed in pancreatitis, as well as PDA, suggested that Id3 might induce cell-cycle entry in ducts. To test this hypothesis, primary human pancreatic duct cells were transduced with an adenovirus-expressing Id3. Remarkably, Id3 expression alone was sufficient to trigger efficient cell-cycle entry, as manifested by expression of the proliferation markers Ki67, phospho-cyclin E, and phospho-histone H3. Collectively, the data establish dysregulation of the Id/bHLH axis as an early and sustained feature of ductal pathogenesis and mark this axis as a potential therapeutic target for intervention in pancreatitis and PDA.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Proteínas de Neoplasias/metabolismo , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Factor de Transcripción 3/metabolismo , Animales , Carcinoma Ductal Pancreático/patología , Ciclo Celular/genética , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Pancreatitis/metabolismo
18.
Aging (Albany NY) ; 2(11): 785-90, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21068465

RESUMEN

Beta-cell replication dramatically declines with age. Here, we report that the level of CENP-A, a protein required for cell division, declines precipitously with age in an islet-specific manner. CENP-A is essentially undetectable after age 29 in humans. However, exocrine cells retain CENP-A expression. The decline in islet-cell CENP-A expression is more striking in humans than in mice, where CENP-A expression continues to be detectable at low levels even in elderly mice. The mechanism by which CENP-A declines appears to be post-transcriptional, as there was no correlation between CENP-A mRNA levels and age or islet purity. This finding has implications for efforts to induce beta-cell replication as a treatment for diabetes.


Asunto(s)
Envejecimiento/fisiología , Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Islotes Pancreáticos/metabolismo , Mitosis , Páncreas Exocrino/citología , Adulto , Animales , Autoantígenos/genética , Proteína A Centromérica , Proteínas Cromosómicas no Histona/genética , Feto/anatomía & histología , Humanos , Ratones , ARN Mensajero/metabolismo
19.
Rev Diabet Stud ; 7(2): 124-31, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21060971

RESUMEN

Beta-cell deficit is the major pathological feature in type 1 and type 2 diabetes patients, and plays a key role in disease progression. In principle, beta-cell regeneration can occur by replication of pre-existing beta-cells, or by beta-cell neogenesis from stem/progenitors. Unfortunately, beta-cell replication is limited by the almost complete absence of beta-cells in patients with type 1 diabetes, and the increasing recognition that the beta-cell replicative capacity declines severely with age. Therefore, beta-cell neogenesis has received increasing interest. Many different cell types within the pancreas have been suggested as potential beta-cell stem/progenitor cells, but the data have been conflicting. In some cases, this may be due to different regeneration models. On the other hand, different results have been obtained with similar regeneration models, leading to confusion about the nature and existence of beta-cell neogenesis in adult animals. Here, we review the major candidates for adult regeneration pathways, and focus on the recent discovery that alpha-cells can function as a novel beta-cell progenitor. Of note, this is a pathway that appears to be unique to beta-cell neogenesis in the adult, as the embryonic pathway of beta-cell neogenesis does not proceed through a glucagon-positive intermediate. We conclude that beta-cell neogenesis from alpha-cells is a new pathway of potential therapeutic significance, making it of high importance to elucidate the molecular events in alpha- to beta-cell conversion.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular , Diabetes Mellitus/terapia , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Animales , Humanos , Células Secretoras de Insulina/trasplante , Trasplante de Células Madre
20.
Stem Cells ; 28(9): 1630-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20653050

RESUMEN

Because type 1 and type 2 diabetes are characterized by loss of ß-cells, ß-cell regeneration has garnered great interest as an approach to diabetes therapy. Here, we developed a new model of ß-cell regeneration, combining pancreatic duct ligation (PDL) with elimination of pre-existing ß-cells with alloxan. In this model, in which virtually all ß-cells observed are neogenic, large numbers of ß-cells were generated within 2 weeks. Strikingly, the neogenic ß-cells arose primarily from α-cells. α-cell proliferation was prominent following PDL plus alloxan, providing a large pool of precursors, but we found that ß-cells could form from α-cells by direct conversion with or without intervening cell division. Thus, classical asymmetric division was not a required feature of the process of α- to ß-cell conversion. Intermediate cells coexpressing α-cell- and ß-cell-specific markers appeared within the first week following PDL plus alloxan, declining gradually in number by 2 weeks as ß-cells with a mature phenotype, as defined by lack of glucagon and expression of MafA, became predominant. In summary, these data revealed a novel function of α-cells as ß-cell progenitors. The high efficiency and rapidity of this process make it attractive for performing the studies required to gain the mechanistic understanding of the process of α- to ß-cell conversion that will be required for eventual clinical translation as a therapy for diabetes.


Asunto(s)
Proliferación Celular , Transdiferenciación Celular , Diabetes Mellitus Experimental/patología , Células Secretoras de Glucagón/patología , Células Secretoras de Insulina/patología , Regeneración , Factores de Edad , Animales , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/metabolismo , Ligadura , Factor de Transcripción MafB/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Proteínas Oncogénicas/metabolismo , Conductos Pancreáticos/cirugía , Fenotipo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...