Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 63(12)2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31570392

RESUMEN

Proteus mirabilis is a common pathogen of the catheterised urinary tract and often described as intrinsically resistant to the biocide chlorhexidine (CHD). Here we demonstrate that de-repression of the smvA efflux system has occurred in clinical isolates of P. mirabilis and reduces susceptibility to CHD and other cationic biocides. Compared to other isolates examined, P. mirabilis RS47 exhibited a significantly higher CHD MIC (≥512 µg/ml) and significantly greater expression of smvA. Comparison of the RS47 smvA and cognate smvR repressor with sequences from other isolates, indicated that RS47 encodes an inactivated smvR. Complementation of RS47 with a functional smvR from isolate RS50a (which exhibited the lowest smvA expression and lowest CHD MIC) reduced smvA expression by ∼59-fold, and markedly lowered the MIC of CHD and other cationic biocides. Although complementation of RS47 did not reduce MICs to concentrations observed in isolate RS50a, the significantly lower polymyxin B MIC of RS50a indicated that differences in LPS structure are also a factor in P. mirabilis CHD susceptibility. To determine if exposure to CHD can select for mutations in smvR, clinical isolates with the lowest CHD MICs were adapted to grow at increasing concentrations of CHD up to 512 µg/ml. Analysis of the smvR in adapted populations indicated that mutations predicted to inactivate smvR occurred following CHD exposure in some isolates. Collectively, our data show that smvA de-repression contributes to reduced biocide susceptibility in P. mirabilis, but differences in LPS structure between strains are also likely to be an important factor.

2.
Lett Appl Microbiol ; 68(4): 277-293, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30811615

RESUMEN

Urethral catheters are the most commonly deployed medical devices and used to manage a wide range of conditions in both hospital and community care settings. The use of long-term catheterization, where the catheter remains in place for a period >28 days remains common, and the care of these patients is often undermined by the acquisition of infections and formation of biofilms on catheter surfaces. Particular problems arise from colonization with urease-producing species such as Proteus mirabilis, which form unusual crystalline biofilms that encrust catheter surfaces and block urine flow. Encrustation and blockage often lead to a range of serious clinical complications and emergency hospital referrals in long-term catheterized patients. Here we review current understanding of bacterial biofilm formation on urethral catheters, with a focus on crystalline biofilm formation by P. mirabilis, as well as approaches that may be used to control biofilm formation on these devices. SIGNIFICANCE AND IMPACT OF THE STUDY: Urinary catheters are the most commonly used medical devices in many healthcare systems, but their use predisposes to infection and provide ideal conditions for bacterial biofilm formation. Patients managed by long-term urethral catheterization are particularly vulnerable to biofilm-related infections, with crystalline biofilm formation by urease producing species frequently leading to catheter blockage and other serious clinical complications. This review considers current knowledge regarding biofilm formation on urethral catheters, and possible strategies for their control.


Asunto(s)
Catéteres de Permanencia/microbiología , Proteus mirabilis/crecimiento & desarrollo , Ureasa/uso terapéutico , Cateterismo Urinario/efectos adversos , Catéteres Urinarios/microbiología , Biopelículas/crecimiento & desarrollo , Humanos , Terapia de Fagos/métodos , Infecciones por Proteus , Proteus mirabilis/patogenicidad , Infecciones Urinarias/microbiología , Infecciones Urinarias/prevención & control
3.
J Endocrinol ; 196(1): 33-43, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18180315

RESUMEN

Generation of new beta-cells from the adult pancreas or the embryonic stem cells is being pursued by research groups worldwide. Success will be dependent on confirmation of true beta-cell phenotype evidenced by capacity to process and store proinsulin. The aim of these studies was to robustly determine endocrine characteristics of the AR42J rat pancreatic acinar cell line before and after in vitro transdifferentiation. beta-cell phenotypic marker expression was characterised by RT-PCR, immunostaining, western blotting, ELISA and in human preproinsulin transgene over-expression studies in wild-type AR42J cells and after culture on Matrigel basement membrane matrix with and without growth/differentiation factor supplementation. Pancreatic duodenal homeobox 1 (PDX1), forkhead box transcription factor a2 (Foxa2), glucokinase, pancreatic polypeptide and low-level insulin gene transcription in wild-type AR42J cells were confirmed by RT-PCR. Culture on Matrigel-coated plates and supplementation of medium with glucagon-like peptide 1 induced expression of the beta-cell Glut 2 with maintained expression of insulin and PDX1. Increased biosynthesis and secretion of proinsulin were confirmed by immunocytochemical staining and sensitive ELISA. Absence of the regulated secretory pathway was demonstrated by undetectable prohormone convertase expression. In addition, inability to process and store endogenous proinsulin or human proinsulin translated from a constitutively over-expressed preproinsulin transgene was confirmed. The importance of robust phenotypic characterisation at the protein level in attempted beta-cell transdifferentiation studies has been confirmed. Rodent and human sensitive/specific differential proinsulin/insulin ELISA in combination with human preproinsulin over-expression enables detailed elucidatation of core endocrine functions of proinsulin processing and storage in putative new beta-cells.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Proinsulina/metabolismo , Animales , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Perfilación de la Expresión Génica , Péptido 1 Similar al Glucagón/farmacología , Glucoquinasa/genética , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Insulina , Células Secretoras de Insulina/química , Masculino , Polipéptido Pancreático/genética , Fenotipo , Proinsulina/biosíntesis , Proinsulina/genética , Precursores de Proteínas/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transfección , Transgenes/genética
4.
Biochem Biophys Res Commun ; 353(4): 1011-6, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17210120

RESUMEN

Recent studies have identified a positive role for nitric oxide (NO) in the regulation of pancreatic beta-cell function. The aim of this study was to determine the effects of short-term exposure to NO on beta-cell gene expression and the activity of the transcription factor PDX-1. NO stimulated the activity of the insulin gene promoter in Min6 beta-cells and endogenous insulin mRNA levels in both Min6 and isolated islets of Langerhans. Addition of wortmannin prior to NO stimulation blocked the observed increases in insulin gene promoter activity. Although NO addition stimulated the phosphorylation of p38, inhibition by SB203580 did not block the effect of NO on the insulin gene promoter. NO addition also stimulated both the nuclear accumulation and the DNA binding activity of PDX-1. This study has shown that over 24h, NO stimulates insulin gene expression, PI-3-kinase activity and the activity of the critical beta-cell transcription factor PDX-1.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Insulina/genética , Óxido Nítrico/farmacología , Transcripción Genética/efectos de los fármacos , Androstadienos/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Humanos , Técnicas In Vitro , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo , Transfección , Wortmanina , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Diabetologia ; 49(4): 685-96, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16489446

RESUMEN

AIMS/HYPOTHESIS: Rosiglitazone and metformin are two oral antihyperglycaemic drugs used to treat type 2 diabetes. While both drugs have been shown to improve insulin-sensitive glucose uptake, the direct effects of these drugs on pancreatic beta cells is only now beginning to be clarified. The aim of the present study was to determine the direct effects of these agents on beta cell gene expression. METHODS: We used reporter gene analysis to examine the effects of rosiglitazone and metformin on the activity of the proinsulin and insulin promoter factor 1 (IPF1) gene promoters in the glucose-responsive mouse beta cell line Min6. Western blot and gel retardation analyses were used to examine the effects of both drugs on the regulation of IPF1 protein production, nuclear accumulation and DNA binding activity in both Min6 cells and isolated rat islets of Langerhans. RESULTS: Over 24 h, rosiglitazone promoted the nuclear accumulation of IPF1 and forkhead homeobox A2 (FOXA2), independently of glucose concentration, and stimulated a two-fold increase in the activity of the Ipf1 gene promoter (p<0.01). Stimulation of the Ipf1 promoter by rosiglitazone was unaffected by the presence of the peroxisome proliferator activated receptor gamma antagonist GW9662. No effect of either rosiglitazone or metformin was observed on proinsulin promoter activity. Metformin stimulated IPF1 nuclear accumulation and DNA binding activity in a time-dependent manner, with maximal effects observed after 2 h. CONCLUSIONS/INTERPRETATION: Metformin and rosiglitazone have direct effects on beta cell gene expression, suggesting that these agents may play a previously unrecognised role in the direct regulation of pancreatic beta cell function.


Asunto(s)
Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Metformina/farmacología , Tiazolidinedionas/farmacología , Transporte Activo de Núcleo Celular , Anilidas/farmacología , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , ADN/metabolismo , Glucosa/farmacología , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proinsulina/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Ratas , Rosiglitazona , Factores de Tiempo , Transactivadores/genética , Transactivadores/metabolismo
6.
Diabetologia ; 44(2): 249-58, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11270685

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to examine the putative role of mutations in the insulin promoter 1 (IPF1) gene in early-onset diabetes. METHODS: We carried out mutation screening of the IPF1 gene in 115 Scandinavian families with at least two members with onset of diabetes younger than 40 years. The allele frequencies were also tested in 183 unrelated patients with late-onset Type II (non-insulin-dependent) diabetes mellitus and in 92 non-diabetic control subjects. RESULTS: Two novel IPF1 variants (G212R and P239Q) and one previously reported (D76N) IPF1 variant were identified in the 115 families (3.5%). The D76N variant was found in one MODY3 family (S315fsinsA of HNF1alpha) and also in two families with late-onset Type II diabetes. The P239Q variant was identified in two families with early-onset diabetes including one with MODY3 (R272C of HNF1alpha) and in three families with late-onset Type II diabetes. Despite the fact that the variants did not segregate completely with diabetes, the non-diabetic carriers of the IPF1 variants had increased blood glucose concentrations (p < 0.05) and reduced insulin:glucose ratios (p < 0.05) during an oral glucose tolerance test compared with non-diabetic family members without these variants. In addition, when the G212R and P239Q variants were expressed in cells without IPF1 i.e.. Nes2y cells, both variants showed about a 50% reduction in their ability to activate insulin gene transcription compared to wild-type IPF1, as measured by reporter gene assay. CONCLUSION/INTERPRETATION: Although mutations in the IPF-1 gene are rare in early- (3.5 %) and late-onset (2.7 % ) Type II diabetes, they are functionally important and occur also in families with other MODY mutations.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Mutación , Transactivadores/genética , Anciano , Alelos , Glucemia/análisis , Presión Sanguínea , Western Blotting , Índice de Masa Corporal , Colesterol/sangre , Análisis Mutacional de ADN , Ayuno , Femenino , Frecuencia de los Genes , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Proteínas de Homeodominio/genética , Humanos , Cinética , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Fenotipo , Países Escandinavos y Nórdicos , Triglicéridos/sangre
7.
Mol Pathol ; 53(1): 1-7, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10884914

RESUMEN

The transcription of genes could be defined as the intricate molecular manoeuvres occurring in the nuclei of cells, which allow the translation of genetic information held in the DNA into the proteins required for life. Gene transcription is the dominant control point in the production of any protein, and is initiated and regulated through the combined activities of a highly specialised set of nuclear proteins. This review examines the role of these protein "transcription factors" in the production of messenger RNA, the information intermediary produced in the nucleus, and transferred to the cytoplasm to serve as a template for protein synthesis. In combination with RNA polymerase, an extraordinary and complex enzyme required to synthesise new RNA molecules, a multitude of transcription factors combine their activities to orchestrate and control this elegant process.


Asunto(s)
ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , Transcripción Genética/fisiología , Elementos de Facilitación Genéticos/fisiología , Humanos , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción/fisiología
8.
Clin Exp Immunol ; 121(1): 100-5, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10886245

RESUMEN

Type 1 diabetes is a T cell-mediated autoimmune disease where a number of islet beta-cell target autoantigens have been characterized on the basis of reactivity with autoantibodies. Nevertheless, there remains uncertainty of the nature of another group of autoantigens associated with the secretory granule fraction of islet beta-cells that appear to be targeted predominantly by autoreactive T cells. We have previously characterized CD4+, HLA-DR-restricted T cell lines from new onset type 1 diabetic patients that are specific for the secretory granule fraction of rat tumour insulinoma, RIN. The T cell line from the first patient, HS, proliferates in response to crude microsomal membranes prepared from a recently established, pure human islet beta-cell line NES2Y. In addition, the HS line also responds to secretory granule fractions prepared from a murine tumour insulinoma grown in RIP-Tag mice, showing the recognition of species-conserved antigen(s) in beta-cells. Using partially matched antigen-presenting cells, the HS T cells and another line derived from a second patient, MR, were shown to be restricted by disease-associated DRB1*0101 and DRB1*0404 alleles, respectively. Neither the HS or MR T cell lines proliferate in response to a large panel of candidate islet cell antigens, including insulin, proinsulin, glutamic acid decarboxylase, the protein tyrosine phosphatase IA-2/phogrin, imogen-38, ICA69 or hsp60. Our data provide compelling evidence of the presence of a group of antigens associated with the secretory granule fraction of islet beta-cells recognized by the T cell lines, whose definition may contribute to our knowledge of disease induction as well as to diagnosis.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Gránulos Citoplasmáticos , Humanos , Islotes Pancreáticos/citología , Ratones , Ratas , Ratas Wistar , Células Tumorales Cultivadas
10.
Diabetes ; 49(6): 953-60, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10866047

RESUMEN

NES2Y is a proliferating human insulin-secreting cell line that we have derived from a patient with persistent hyperinsulinemic hypoglycemia of infancy. This disease is characterized by unregulated insulin release despite profound hypoglycemia. NES2Y cells, like beta-cells isolated from the patient of origin, lack functional ATP-sensitive potassium channels (KATP) and also carry a defect in the insulin gene-regulatory transcription factor PDX1. Here, we report that the NES2Y beta-cells that are transfected with the genes encoding the components of KATP channels in beta-cells, sulfonylurea receptor (SUR) 1 and Kir6.2, have operational KATP channels and show normal intracellular Ca2+ and secretory responses to glucose. However, these cells, designated NESK beta-cells, have impaired insulin gene transcription responses to glucose. NES2Y beta-cells that are transfected with either Kir6.2 or SUR1 alone do not express functional KATP channels and have impaired intracellular free Ca2+ concentration-signaling responses to depolarization-dependent beta-cell agonists. These findings document that in NES2Y beta-cells, coexpression of both subunits is critically required for fully operational KATP channels and KATP channel-dependent signaling events. This article further characterizes the properties of the novel human beta-cell line, NES2Y, and documents the usefulness of these cells in diabetes-related research.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Canales de Potasio de Rectificación Interna , Canales de Potasio/metabolismo , Receptores de Droga/metabolismo , Calcio/metabolismo , Señalización del Calcio , Línea Celular , Electrofisiología , Humanos , Insulina/genética , Secreción de Insulina , Membranas Intracelulares/metabolismo , Islotes Pancreáticos/fisiología , Concentración Osmolar , Canales de Potasio/genética , Receptores de Sulfonilureas , Transcripción Genética , Transfección
11.
Diabetes ; 49(3): 418-23, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10868963

RESUMEN

Glucose regulates insulin production in pancreatic beta-cells in the long term by stimulating insulin gene transcription. These effects are partially mediated through the activity of a homeodomain transcription factor, PDX-1, which binds to four sites within the human insulin gene promoter. The availability of a human beta-like cell line, NES2Y, which lacks PDX-1 but expresses the insulin gene, allowed us to determine whether PDX-1 was essential for the stimulatory effect of glucose on insulin mRNA levels. In NES2Y cells, glucose had no effect on the insulin gene promoter linked to a firefly luciferase reporter or on endogenous insulin mRNA levels. However, in NES2Y cells stably transfected with PDX-1 (NES-PDX-1), glucose exhibited a marked stimulatory effect on both the insulin promoter (5+/-0.2-fold, n = 6) and insulin mRNA levels (4.8+/-0.5-fold, n = 4). NES2Y cells were derived from a patient with persistent hyperinsulinemic hypoglycemia of infancy; the cells therefore lacked operational ATP-sensitive potassium channels, which results in the failure to control depolarization-dependent intracellular Ca2+ signaling. Despite the loss of control of Ca2+ channel activity, NES-PDX-1 cells maintained normal glucose-responsive insulin gene regulation. These results demonstrate that glucose modulation of insulin mRNA levels is dependent on the activity of PDX-1 and that these effects are independent of changes in intracellular Ca2+ concentrations.


Asunto(s)
Calcio/metabolismo , Glucosa/farmacología , Proteínas de Homeodominio , Insulina/genética , Membranas Intracelulares/metabolismo , ARN Mensajero/metabolismo , Transactivadores/fisiología , Línea Celular , Humanos , Concentración Osmolar , Enfermedades Pancreáticas/metabolismo , Enfermedades Pancreáticas/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Transactivadores/genética , Transfección
12.
J Biol Chem ; 275(20): 15330-5, 2000 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-10748090

RESUMEN

Islet amyloid polypeptide (IAPP) and insulin are expressed in the beta-cells of the islets of Langerhans. They are co-secreted in response to changes in glucose concentration, and their mRNA levels are also regulated by glucose. The promoters of both genes share similar cis-acting sequence elements, and both bind the homeodomain transcription factor PDX1, which plays an important role in the regulation of the insulin promoter and insulin mRNA levels by glucose. Here we examine the role of PDX1 in the regulation of the human IAPP promoter by glucose. The experiments were facilitated by the availability of a human beta-cell line (NES2Y) that lacks PDX1. NES2Y cells also lack operational K(ATP) channels, resulting in a loss of control of calcium signaling. We have previously used these cells to show that glucose regulation of the insulin gene is dependent on PDX1, but not calcium. In the mouse beta-cell line Min6, glucose (16 mm) stimulated a 3.5-4-fold increase in the activity of a -222 to +450 IAPP promoter construct compared with values observed in 0.5 mm glucose. In NES2Y cells, glucose failed to stimulate transcriptional activation of the IAPP promoter. Overexpression of PDX1 in NES2Y cells failed to reinstate glucose-responsive control of the IAPP promoter. Glucose effects on the IAPP promoter were observed only in the presence of PDX1 when normal calcium signaling was restored by overexpression of the two K(ATP) channel subunits SUR1 and Kir6.2. The importance of calcium was further emphasized by an experiment in which glucose-stimulated IAPP promoter activity was inhibited by the calcium channel blocker verapamil (50 microm). Verapamil was further shown to inhibit the stimulatory effect of glucose on IAPP mRNA levels. These results demonstrate that like the insulin promoter, glucose regulation of the IAPP promoter is dependent on the activity of PDX1, but unlike the insulin promoter, it additionally requires the activity of another, as yet uncharacterized factor(s), the activity of which is calcium-dependent.


Asunto(s)
Amiloide/genética , Calcio/fisiología , Glucosa/farmacología , Islotes Pancreáticos/metabolismo , Transactivadores/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Calcio/farmacología , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Polipéptido Amiloide de los Islotes Pancreáticos , Islotes Pancreáticos/citología , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/genética , Proteínas Recombinantes/biosíntesis , Transactivadores/deficiencia , Transactivadores/genética , Transfección , Verapamilo/farmacología
13.
FEBS Lett ; 463(1-2): 53-7, 1999 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-10601637

RESUMEN

Pax4 is a paired-box transcription factor that plays an important role in the development of pancreatic beta-cells. Two Pax4 cDNAs were isolated from a rat insulinoma library. One contained the full-length sequence of Pax4. The other, termed Pax4c, was identical to Pax4 but lacked the sequences encoding 117 amino acids at the COOH-terminus. Pax4 was found to inhibit the human insulin promoter through a sequence element, the C2 box, located at -253 to -244, and the islet amyloid polypeptide promoter through a sequence element located downstream of -138. The inhibitory activity of Pax4 was mapped to separate regions of the protein between amino acids 2-230 and 231-349.


Asunto(s)
Amiloide/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Antagonistas de Insulina , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Amiloide/genética , Animales , Línea Celular , Clonación Molecular , ADN Complementario/metabolismo , Biblioteca de Genes , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Ratones , Factores de Transcripción Paired Box , Plásmidos , Biosíntesis de Proteínas , Isoformas de Proteínas , Ratas , Transfección
14.
Biochem J ; 344 Pt 3: 813-8, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10585868

RESUMEN

Pancreatic-duodenal homoeobox factor-1 (PDX1) is a homoeodomain transcription factor that plays an important role in linking glucose metabolism in pancreatic beta cells to the regulation of insulin gene transcription. Our previous results indicated that glucose activates PDX1 DNA-binding activity and insulin promoter activity via a stress-activated signalling pathway involving phosphatidylinositol 3-kinase (PtdIns 3-kinase) and stress-activated protein kinase 2 (SAPK2/p38). The present study was undertaken to determine the effects of other metabolizable and non-metabolizable nutrients. The results indicate that non-metabolizable nutrients, with the exception of 2-deoxyglucose, had no effect. Metabolizable nutrients that could stimulate calcium uptake and insulin release were shown to activate both PDX1 and the insulin promoter. The possible role of insulin acting via an autoregulatory loop was therefore examined. Insulin was shown to potently activate PDX1 DNA-binding activity and insulin promoter activity. The effects of insulin were inhibited by the PtdIns 3-kinase inhibitors wortmannin and LY294002 and by the SAPK2 inhibitor SB203580, suggesting that its effects were mediated via activation of PtdIns 3-kinase and SAPK2. Further support for the insulin-mediated activation of SAPK2 came from the observation that both glucose and insulin stimulated the phosphorylation of SAPK2. These results suggest that both glucose and insulin stimulate PDX1 DNA-binding activity and insulin promoter activity via a pathway involving PtdIns 3-kinase and SAPK2.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Insulina/farmacología , Islotes Pancreáticos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Androstadienos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Cromonas/farmacología , Glucosa/farmacología , Humanos , Imidazoles/farmacología , Islotes Pancreáticos/metabolismo , Morfolinas/farmacología , Oligodesoxirribonucleótidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Piridinas/farmacología , Transfección , Wortmanina , Proteínas Quinasas p38 Activadas por Mitógenos
15.
J Clin Invest ; 104(9): R33-9, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10545530

RESUMEN

The transcription factor insulin promoter factor-1 (IPF-1) plays a central role in both the development of the pancreas and the regulation of insulin gene expression in the mature pancreatic beta cell. A dominant-negative frameshift mutation in the IPF-l gene was identified in a single family and shown to cause pancreatic agenesis when homozygous and maturity-onset diabetes of the young (MODY) when heterozygous. We studied the role of IPF-1 in Caucasian diabetic and nondiabetic subjects from the United Kingdom. Three novel IPF-1 missense mutations (C18R, D76N, and R197H) were identified in patients with type 2 diabetes. Functional analyses of these mutations demonstrated decreased binding activity to the human insulin gene promoter and reduced activation of the insulin gene in response to hyperglycemia in the human beta-cell line Nes2y. These mutations are present in 1% of the population and predisposed the subject to type 2 diabetes with a relative risk of 3.0. They were not highly penetrant MODY mutations, as there were nondiabetic mutation carriers 25-53 years of age. We conclude that mutations in the IPF-1 gene may predispose to type 2 diabetes and are a rare cause of MODY and pancreatic agenesis, with the phenotype depending upon the severity of the mutation.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodominio , Transactivadores/genética , Adulto , Anciano , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Glucosa/metabolismo , Humanos , Insulina/genética , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Fenotipo , Fosforilación , Transcripción Genética
16.
J Biol Chem ; 274(48): 34059-66, 1999 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-10567373

RESUMEN

Persistent hyperinsulinemic hypoglycemia of infancy (PHHI) is a neonatal disease characterized by dysregulation of insulin secretion accompanied by profound hypoglycemia. We have discovered that islet cells, isolated from the pancreas of a PHHI patient, proliferate in culture while maintaining a beta cell-like phenotype. The PHHI-derived cell line (NES2Y) exhibits insulin secretory characteristics typical of islet cells derived from these patients, i.e. they have no K(ATP) channel activity and as a consequence secrete insulin at constitutively high levels in the absence of glucose. In addition, they exhibit impaired expression of the homeodomain transcription factor PDX1, which is a key component of the signaling pathway linking nutrient metabolism to the regulation of insulin gene expression. To repair these defects NES2Y cells were triple-transfected with cDNAs encoding the two components of the K(ATP) channel (SUR1 and Kir6.2) and PDX1. One selected clonal cell line (NISK9) had normal K(ATP) channel activity, and as a result of changes in intracellular Ca(2+) homeostasis ([Ca(2+)](i)) secreted insulin within the physiological range of glucose concentrations. This approach to engineering PHHI-derived islet cells may be of use in gene therapy for PHHI and in cell engineering techniques for administering insulin for the treatment of diabetes mellitus.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Glucosa/farmacología , Proteínas de Homeodominio , Hiperinsulinismo/genética , Hipoglucemia/genética , Insulina/metabolismo , Islotes Pancreáticos/citología , Canales de Potasio de Rectificación Interna , Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Animales , Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Electrofisiología , Ingeniería Genética , Humanos , Hiperinsulinismo/patología , Hipoglucemia/patología , Lactante , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Canales de Potasio/genética , Cloruro de Potasio/farmacología , Receptores de Droga/genética , Proteínas Recombinantes de Fusión/genética , Receptores de Sulfonilureas , Tolbutamida/farmacología , Transactivadores/genética , Transfección , Células Tumorales Cultivadas
17.
J Biol Chem ; 274(2): 1011-6, 1999 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-9873045

RESUMEN

One of the mechanisms whereby glucose stimulates insulin gene transcription in pancreatic beta-cells involves activation of the homeodomain transcription factor PDX1 (pancreatic/duodenal homeobox-1) via a stress-activated pathway involving stress-activated protein kinase 2 (SAPK2, also termed RK/p38, CSBP, and Mxi2). In the present study we show, by Western blotting and electrophoretic mobility shift assay, that in human islets of Langerhans incubated in low glucose (3 mM) PDX1 exists as an inactive 31-kDa protein localized exclusively in the cytoplasm. Transfer of the islets to high (16 mM) glucose results in rapid (within 10 min) conversion of PDX1 to an active 46-kDa form that was present predominantly in the nucleus. Activation of PDX1 appears to involve phosphorylation, as shown by incorporation of 32Pi into the 46-kDa form of the protein. These effects of glucose could be mimicked by chemical stress (sodium arsenite), or by overexpression of SAPK2 in the beta-cell line MIN6. Overexpression of SAPK2 also stimulated PDX1-dependent transcription of a -50 to -250 region of the human insulin gene promoter linked to a firefly luciferase reporter gene. The effects of glucose were inhibited by the SAPK2 inhibitor SB 203580, and by wortmannin and LY 294002, which inhibit phosphatidylinositol 3-kinase, although the effects of stress (arsenite) were inhibited only by SB 203580. These results demonstrate that glucose regulates the insulin gene promoter through activation and nuclear translocation of PDX1 via the SAPK2 pathway.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Glucosa/farmacología , Proteínas de Homeodominio/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos , Transactivadores/metabolismo , Secuencia de Bases , Transporte Biológico , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Cartilla de ADN , Inhibidores Enzimáticos/farmacología , Humanos , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos
18.
J Biol Chem ; 272(33): 20936-44, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9252422

RESUMEN

Insulin upstream factor 1 (IUF1), a transcription factor present in pancreatic beta-cells, binds to the sequence C(C/T)TAATG present at several sites within the human insulin promoter. Here we isolated and sequenced cDNA encoding human IUF1 and exploited it to identify the signal transduction pathway by which glucose triggers its activation. In human islets, or in the mouse beta-cell line MIN6, high glucose induced the binding of IUF1 to DNA, an effect mimicked by serine/threonine phosphatase inhibitors, indicating that DNA binding was induced by a phosphorylation mechanism. The glucose-stimulated binding of IUF1 to DNA and IUF1-dependent gene transcription were both prevented by SB 203580, a specific inhibitor of stress-activated protein kinase 2 (SAPK2, also termed p38 mitogen-activated protein kinase, reactivating kinase, CSBP, and Mxi2) but not by several other protein kinase inhibitors. Consistent with this finding, high glucose activated mitogen-activated protein kinase-activated protein kinase 2 (MAPKAP kinase-2) (a downstream target of SAPK2) in MIN6 cells, an effect that was also blocked by SB 203580. Cellular stresses that trigger the activation of SAPK2 and MAPKAP kinase-2 (arsenite, heat shock) also stimulated IUF1 binding to DNA and IUF1-dependent gene transcription, and these effects were also prevented by SB 203580. IUF1 expressed in Escherichia coli was unable to bind to DNA, but binding was induced by incubation with MgATP, SAPK2, and a MIN6 cell extract, which resulted in the conversion of IUF1 to a slower migrating form. SAPK2 could not be replaced by p42 MAP kinase, MAPKAP kinase-2, or MAPKAP kinase-3. The glucose-stimulated activation of IUF1 DNA binding and MAPKAP kinase-2 (but not the arsenite-induced activation of these proteins) was prevented by wortmannin and LY 294002 at concentrations similar to those that inhibit phosphatidylinositide 3-kinase. Our results indicate that high glucose (a cellular stress) activates SAPK2 by a novel mechanism in which a wortmannin/LY 294002-sensitive component plays an essential role. SAPK2 then activates IUF1 indirectly by activating a novel IUF1-activating enzyme.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Glucosa/farmacología , Insulina/genética , Islotes Pancreáticos/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Factores de Transcripción/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Clonación Molecular , ADN/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Fosfatidilinositol 3-Quinasas , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
19.
FEBS Lett ; 413(2): 304-8, 1997 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-9280302

RESUMEN

Persistent hyperinsulinaemic hypoglycaemia of infancy (PHHI), or nesidioblastosis, is a rare disorder which may be familial or sporadic, and which is characterized by unregulated secretion of insulin and profound hypoglycaemia in the neonate. The defect has been linked in some patients to mutations in the sulphonyl urea receptor gene (SUR). The present study investigated potential defects in the regulation of the insulin gene by glucose in a beta-cell line (NES 2Y) derived from a patient with PHHI. The results show that the insulin promoter is unresponsive to glucose in PHHI, and that this defect can be attributed to impaired expression of the transcription factor IUF1. Because IUF1 is involved not only in linking glucose metabolism to the control of the insulin, but is also a major regulator of beta-cell differentiation during embryogenesis, we propose that impaired expression of IUF1 contributes to beta-cell dysfunction in PHHI by leading to abnormal beta-cell differentiation.


Asunto(s)
Proteínas de Unión al ADN , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio , Insulina/genética , Islotes Pancreáticos/fisiología , Enfermedades Pancreáticas/genética , Factores de Transcripción/genética , Línea Celular , ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Proteínas Recombinantes de Fusión , Transactivadores/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Factores Estimuladores hacia 5'
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...