Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci Alliance ; 7(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38876803

RESUMEN

A lack of social relationships is increasingly recognized as a type 2 diabetes (T2D) risk. To investigate the underlying mechanism, we used male KK mice, an inbred strain with spontaneous diabetes. Given the association between living alone and T2D risk in humans, we divided the non-diabetic mice into singly housed (KK-SH) and group-housed control mice. Around the onset of diabetes in KK-SH mice, we compared H3K27ac ChIP-Seq with RNA-Seq using pancreatic islets derived from each experimental group, revealing a positive correlation between single-housing-induced changes in H3K27ac and gene expression levels. In particular, single-housing-induced H3K27ac decreases revealed a significant association with islet cell functions and GWAS loci for T2D and related diseases, with significant enrichment of binding motifs for transcription factors representative of human diabetes. Although these H3K27ac regions were preferentially localized to a polymorphic genomic background, SNVs and indels did not cause sequence disruption of enriched transcription factor motifs in most of these elements. These results suggest alternative roles of genetic variants in environment-dependent epigenomic changes and provide insights into the complex mode of disease inheritance.


Asunto(s)
Diabetes Mellitus Tipo 2 , Epigenómica , Islotes Pancreáticos , Animales , Ratones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Epigenómica/métodos , Histonas/metabolismo , Polimorfismo de Nucleótido Simple , Epigénesis Genética/genética , Diabetes Mellitus Experimental/genética , Estudio de Asociación del Genoma Completo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
2.
Sci Rep ; 13(1): 17958, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37863964

RESUMEN

The mechanisms of impaired glucose-induced insulin secretion from the pancreatic ß-cells in obesity have not yet been completely elucidated. Here, we aimed to assess the effects of adipocyte-derived factors on the functioning of pancreatic ß-cells. We prepared a conditioned medium using 3T3-L1 cell culture supernatant collected at day eight (D8CM) and then exposed the rat pancreatic ß-cell line, INS-1D. We found that D8CM suppressed insulin secretion in INS-1D cells due to reduced intracellular calcium levels. This was mediated by the induction of a negative regulator of insulin secretion-NECAB1. LC-MS/MS analysis results revealed that D8CM possessed steroid hormones (cortisol, corticosterone, and cortisone). INS-1D cell exposure to cortisol or corticosterone increased Necab1 mRNA expression and significantly reduced insulin secretion. The increased expression of Necab1 and reduced insulin secretion effects from exposure to these hormones were completely abolished by inhibition of the glucocorticoid receptor (GR). NECAB1 expression was also increased in the pancreatic islets of db/db mice. We demonstrated that the upregulation of NECAB1 was dependent on GR activation, and that binding of the GR to the upstream regions of Necab1 was essential for this effect. NECAB1 may play a novel role in the adipoinsular axis and could be potentially involved in the pathophysiology of obesity-related diabetes mellitus.


Asunto(s)
Secreción de Insulina , Células Secretoras de Insulina , Receptores de Glucocorticoides , Animales , Ratones , Ratas , Cromatografía Liquida , Corticosterona/metabolismo , Glucosa/metabolismo , Hidrocortisona/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Obesidad/metabolismo , Receptores de Glucocorticoides/metabolismo , Espectrometría de Masas en Tándem
3.
Bioorg Med Chem ; 31: 115966, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33387694

RESUMEN

Thymidine derivatives bearing spiroacetal moieties on the C4'-position (5'R-spiro-thymidine and 5'S-spiro-thymidine) were synthesized and incorporated into oligonucleotides. The duplex- and triplex-forming abilities of both the oligonucleotides were evaluated from UV melting experiments. Oligonucleotides with the 5'S-spiro modifications could form thermally stable duplexes with complementary RNA and DNA; however, the 5'R-spiro modification significantly decreased the thermal stabilities of the duplexes and triplexes. Oligonucleotides with these spiro-thymidines showed significantly high resistance towards enzymatic degradation.


Asunto(s)
Oligonucleótidos/química , Compuestos de Espiro/química , Timidina/química , Estructura Molecular , Oligonucleótidos/síntesis química
4.
J Diabetes Investig ; 11(4): 814-822, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31957256

RESUMEN

AIMS/INTRODUCTION: Taste receptors, T1rs and T2rs, and the taste-selective G-protein, α-gustducin, are expressed outside the taste-sensing system, such as enteroendocrine L cells. Here, we examined whether α-gustducin also affects nutrition sensing and insulin secretion by pancreatic ß-cells. MATERIALS AND METHODS: The expression of α-gustducin and taste receptors was evaluated in ß-cell lines, and in rat and mouse islets either by quantitative polymerase chain reaction or fluorescence immunostaining. The effects of α-gustducin knockdown on insulin secretion and on cyclic adenosine monophosphate and intracellular Ca2+ levels in rat INS-1 cells were estimated. Sucralose (taste receptor agonist)-induced insulin secretion was investigated in INS-1 cells with α-gustducin suppression and in islets from mouse disease models. RESULTS: The expression of Tas1r3 and α-gustducin was confirmed in ß-cell lines and pancreatic islets. Basal levels of cyclic adenosine monophosphate, intracellular calcium and insulin secretion were significantly enhanced with α-gustducin knockdown in INS-1 cells. The expression of α-gustducin was decreased in high-fat diet-fed mice and in diabetic db/db mice. Sucralose-induced insulin secretion was not attenuated in INS-1 cells with α-gustducin knockdown or in mouse islets with decreased expression of α-gustducin. CONCLUSIONS: α-Gustducin is involved in the regulation of cyclic adenosine monophosphate, intracellular calcium levels and insulin secretion in pancreatic ß-cells in a manner independent of taste receptor signaling. α-Gustducin might play a novel role in ß-cell physiology and the development of type 2 diabetes.


Asunto(s)
Secreción de Insulina/fisiología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Transducina/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Humanos , Proteínas Sensoras del Calcio Intracelular/metabolismo , Ratones , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
5.
Diabetologia ; 61(12): 2608-2620, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30284014

RESUMEN

AIMS/HYPOTHESIS: Epigenetic regulation of gene expression has been implicated in the pathogenesis of obesity and type 2 diabetes. However, detailed information, such as key transcription factors in pancreatic beta cells that mediate environmental effects, is not yet available. METHODS: To analyse genome-wide cis-regulatory profiles and transcriptome of pancreatic islets derived from a diet-induced obesity (DIO) mouse model, we conducted chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-Seq) of histone H3 lysine 27 acetylation (histone H3K27ac) and high-throughput RNA sequencing. Transcription factor-binding motifs enriched in differential H3K27ac regions were examined by de novo motif analysis. For the predicted transcription factors, loss of function experiments were performed by transfecting specific siRNA in INS-1, a rat beta cell line, with and without palmitate treatment. Epigenomic and transcriptional changes of possible target genes were evaluated by ChIP and quantitative RT-PCR. RESULTS: After long-term feeding with a high-fat diet, C57BL/6J mice were obese and mildly glucose intolerant. Among 39,350 islet cis-regulatory regions, 13,369 and 4610 elements showed increase and decrease in ChIP-Seq signals, respectively, significantly associated with global change in gene expression. Remarkably, increased H3K27ac showed a distinctive genomic localisation, mainly in the proximal-promoter regions, revealing enriched elements for nuclear respiratory factor 1 (NRF1), GA repeat binding protein α (GABPA) and myocyte enhancer factor 2A (MEF2A) by de novo motif analysis, whereas decreased H3K27ac was enriched for v-maf musculoaponeurotic fibrosarcoma oncogene family protein K (MAFK), a known negative regulator of beta cells. By siRNA-mediated knockdown of NRF1, GABPA or MEF2A we found that INS-1 cells exhibited downregulation of fatty acid ß-oxidation genes in parallel with decrease in the associated H3K27ac. Furthermore, in line with the epigenome in DIO mice, palmitate treatment caused increase in H3K27ac and induction of ß-oxidation genes; these responses were blunted when NRF1, GABPA or MEF2A were suppressed. CONCLUSIONS/INTERPRETATION: These results suggest novel roles for DNA-binding proteins and fatty acid signalling in obesity-induced epigenomic regulation of beta cell function. DATA AVAILABILITY: The next-generation sequencing data in the present study were deposited at ArrayExpress. RNA-Seq: Dataset name: ERR2538129 (Control), ERR2538130 (Diet-induced obesity) Repository name and number: E-MTAB-6718 - RNA-Seq of pancreatic islets derived from mice fed a long-term high-fat diet against chow-fed controls. ChIP-Seq: Dataset name: ERR2538131 (Control), ERR2538132 (Diet-induced obesity) Repository name and number: E-MTAB-6719 - H3K27ac ChIP-Seq of pancreatic islets derived from mice fed a long-term high-fat diet (HFD) against chow-fed controls.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Acetilación , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Int J Mol Med ; 36(1): 222-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25955334

RESUMEN

Although recent genome-wide association studies (GWAS) have been extremely successful, it remains a big challenge to functionally annotate disease­associated single nucleotide polymorphisms (SNPs), as the majority of these SNPs are located in non­coding regions of the genome. In this study, we described a novel strategy for identifying the proteins that bind to the SNP­containing locus in an allele­specific manner and successfully applied this method to SNPs in the type 2 diabetes mellitus susceptibility gene, potassium voltage­gated channel, KQT­like subfamily Q, member 1 (KCNQ1). DNA fragments encompassing SNPs, and risk or non­risk alleles were immobilized onto the novel nanobeads and DNA­binding proteins were purified from the nuclear extracts of pancreatic ß cells using these DNA­immobilized nanobeads. Comparative analysis of the allele-specific DNA-binding proteins indicated that the affinities of several proteins for the examined SNPs differed between the alleles. Nuclear transcription factor Y (NF­Y) specifically bound the non­risk allele of the SNP rs2074196 region and stimulated the transcriptional activity of an artificial promoter containing SNP rs2074196 in an allele­specific manner. These results suggest that SNP rs2074196 modulates the affinity of the locus for NF­Y and possibly induces subsequent changes in gene expression. The findings of this study indicate that our comparative method using novel nanobeads is effective for the identification of allele­specific DNA­binding proteins, which may provide important clues for the functional impact of disease­associated non­coding SNPs.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus Tipo 2/genética , Canal de Potasio KCNQ1/genética , Nanopartículas de Magnetita , Animales , Línea Celular Tumoral , Predisposición Genética a la Enfermedad , Células HEK293 , Células HeLa , Humanos , Células Secretoras de Insulina/metabolismo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , Ratas , Activación Transcripcional/genética
7.
PLoS One ; 9(8): e104184, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25126749

RESUMEN

The postnatal proliferation and maturation of insulin-secreting pancreatic ß-cells are critical for glucose metabolism and disease development in adults. Elucidation of the molecular mechanisms underlying these events will be beneficial to direct the differentiation of stem cells into functional ß-cells. Maturation of ß-cells is accompanied by increased expression of MafA, an insulin gene transcription factor. Transcriptome analysis of MafA knockout islets revealed MafA is required for the expression of several molecules critical for ß-cell function, including Glut2, ZnT8, Granuphilin, Vdr, Pcsk1 and Urocortin 3, as well as Prolactin receptor (Prlr) and its downstream target Cyclin D2 (Ccnd2). Inhibition of MafA expression in mouse islets or ß-cell lines resulted in reduced expression of Prlr and Ccnd2, and MafA transactivated the Prlr promoter. Stimulation of ß-cells by prolactin resulted in the phosphorylation and translocation of Stat5B and an increased nuclear pool of Ccnd2 via Prlr and Jak2. Consistent with these results, the loss of MafA resulted in impaired proliferation of ß-cells at 4 weeks of age. These results suggest that MafA regulates the postnatal proliferation of ß-cells via prolactin signaling.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Animales , Animales Recién Nacidos , Línea Celular , Proliferación Celular , Ciclina D2/genética , Ciclina D2/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Ratas , Receptores de Prolactina/genética , Transducción de Señal , Transcriptoma
8.
Endocrinology ; 154(11): 4388-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029239

RESUMEN

The luciferase reporter system is useful for the assessment of various biological processes in vivo. The transcription factor pancreatic and duodenal homeobox 1 (Pdx1) is critical for the formation and the function of pancreatic ß-cells. A novel reporter system using secreted Gaussia princeps luciferase (GLuc) under the control of a Pdx1 promoter was generated and activated in rat and mouse ß-cell lines. This Pdx1-GLuc construct was used as a transgene for the generation of reporter mice to monitor Pdx1 promoter activity in vivo via the measurement of secreted GLuc activity in a small aliquot of blood. Significantly increased plasma GLuc activity was observed in Pdx1-GLuc mice. Analysis of Pdx1-GLuc mice by bioluminescence imaging, GLuc reporter assays using homogenates of various organs, and immunohistochemistry revealed that GLuc expression and activity were exponentially higher in pancreatic ß-cells than in pancreatic non-ß-cells, the duodenum, and other organs. In addition, GLuc activity secreted into the culture medium from islets isolated from Pdx1-GLuc mice correlated with the number of islets. The transplantation of Pdx1-GLuc islets into severe combined immunodeficiency mice elevated their plasma GLuc activity. Conversely, a partial pancreatectomy in Pdx1-GLuc mice reduced plasma GLuc activity. These results suggest that a secreted luciferase reporter system in vivo enables not only the monitoring of promoter activity but also a quantitative and minimally invasive assessment of physiological and pathological changes in small cell masses, such as pancreatic ß-cells.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Luciferasas/metabolismo , Regiones Promotoras Genéticas/fisiología , Transactivadores/metabolismo , Animales , Línea Celular , Copépodos/enzimología , Expresión Génica , Genes Reporteros , Proteínas de Homeodominio/genética , Islotes Pancreáticos/metabolismo , Luciferasas/genética , Ratones , Ratas , Transactivadores/genética
9.
J Immunol ; 184(6): 2785-92, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20142360

RESUMEN

IgA is a specific isotype required for mucosal immunity and is the most abundant Ab produced in vivo. Recently, several inductive signals for IgA class switch recombination have been identified; however, the molecular details of the action of these signals and the specific factors acting in B cells remain elusive. In this study, we show that combination of retinoic acid (RA) and TGF-beta1 with other factors induced a much higher frequency of IgA-switched cells than reported previously. In addition, IgA production is severely impaired in Runx2-Runx3 double-deficient mice. In Runx2-Runx3-deficient B cells, both RA- and TGF-beta1-dependent inductions of alpha germline transcription are completely blocked. These data suggest that Runx proteins play an essential role in IgA class switching acting downstream of RA and TGF-beta1 signaling.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Inmunoglobulina A/biosíntesis , Cambio de Clase de Inmunoglobulina , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/fisiología , Tretinoina/fisiología , Animales , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Reordenamiento Génico de Linfocito B/inmunología , Inmunoglobulina A/genética , Cambio de Clase de Inmunoglobulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Recombinación Genética/inmunología , Transducción de Señal/genética
10.
J Biol Chem ; 281(19): 13015-13020, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16543239

RESUMEN

Insulin secretion from pancreatic beta-cells occurs by sequential cellular processes, including glucose metabolism, electrical activity, Ca2+ entry, and regulated exocytosis. Abnormalities in any of these functions can impair insulin secretion. In the present study, we demonstrate that inhibition of proteasome activity severely reduces insulin secretion in the mouse pancreatic beta-cell line MIN6-m9. Although no significant effects on glucose metabolism including ATP production were found in the presence of proteasome inhibitors, both glucose- and KCl-induced Ca2+ entry were drastically reduced. As Ca2+-ionophore-induced insulin secretion was unaffected by proteasome inhibition, a defect in Ca2+ entry through voltage-dependent calcium channels (VDCCs) is the likely cause of the impaired insulin secretion. We found that the pore-forming alpha-subunit of VDCCs undergoes ubiquitination, which does not decrease but slightly increases expression of the alpha-subunit protein at the plasma membrane. However, electrophysiological analysis revealed that treatment with proteasome inhibitors results in a severe reduction in VDCC activity in MIN6-m9 cells, indicating that VDCC function is suppressed by proteasome inhibition. Furthermore, insulin secretion in isolated mouse pancreatic islets was also decreased by proteasome inhibition. These results demonstrate that the ubiquitin-proteasome system plays a critical role in insulin secretion by maintaining normal function of VDCCs.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Línea Celular , Regulación de la Expresión Génica , Glucosa , Secreción de Insulina , Ratones , Cloruro de Potasio , Inhibidores de Proteasoma
11.
Proc Natl Acad Sci U S A ; 102(42): 15116-21, 2005 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-16210247

RESUMEN

Although several studies have suggested that insulin-secreting cells can be generated in vitro from cells residing in adult exocrine pancreas, neither the origin of these cells nor their precise insulin secretory properties was obtained. We show here that insulin-secreting cells can be derived from adult mouse pancreatic exocrine cells by suspension culture in the presence of EGF and nicotinamide. The frequency of insulin-positive cells was only 0.01% in the initial preparation and increased to approximately 5% in the culture conditions. Analysis by the Cre/loxP-based direct cell lineage tracing system indicates that these newly made cells originate from amylase/elastase-expressing pancreatic acinar cells. Insulin secretion is stimulated by glucose, sulfonylurea, and carbachol, and potentiation by glucagon-like peptide-1 also occurs. Insulin-containing secretory granules are present in these cells. In addition, we found that the enzymatic dissociation of pancreatic acini itself leads to activation of EGF signaling, and that inhibition of EGF receptor kinase blocks the transdifferentiation. These data demonstrate that pancreatic acinar cells can transdifferentiate into insulin-secreting cells with secretory properties similar to those of native pancreatic beta cells, and that activation of EGF signaling is required in such transdifferentiation.


Asunto(s)
Linaje de la Célula , Insulina/metabolismo , Páncreas Exocrino/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Quelantes/metabolismo , Ditizona/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Perfilación de la Expresión Génica , Genes Reporteros , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas Exocrino/fisiología , Transducción de Señal/fisiología
12.
Am J Transplant ; 5(8): 1848-55, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15996231

RESUMEN

Although application of the Edmonton protocol has markedly improved outcomes for pancreatic islet transplantation, the insulin independence rate after islet transplantation from one donor pancreas has proven to remain low. During the isolation process and subsequent clinical transplantation, islets are subjected to severe adverse conditions that impair survival and ultimately contribute to graft failure. Pancreas preservation with the two-layer method (TLM) has proven to improve transplant results by protecting isolated islets against apoptosis through the mitochondrial pathway. However, pancreas storage with TLM cannot protect against activation of c-Jun NH2-terminal kinase (JNK) in isolated islets. This study investigated whether delivery of a JNK inhibitory peptide (JNKI) via the protein transduction system can prevent apoptosis of islet cells immediately after isolation. For efficient delivery of the (JNKI into isolated islets, we synthesized JNKI as a C-terminal fusion peptide with the 11-arginine protein transduction domain (11R-JNKI). 11R efficiently delivered the JNKI into isolated islets and 11R-JNKI prevented islet apoptosis immediately after isolation and improved islet graft function. These findings suggest that peptide drugs could be useful for the prevention of the impairment of islet cells and lead to improvement in the outcomes for pancreatic islet transplantation.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia de Injerto/fisiología , Trasplante de Islotes Pancreáticos/fisiología , Islotes Pancreáticos/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Fragmentos de Péptidos/fisiología , Secuencia de Aminoácidos , Animales , Permeabilidad de la Membrana Celular , Frío , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/prevención & control , Ratones , Ratones SCID , Datos de Secuencia Molecular , Preservación de Órganos , Fragmentos de Péptidos/química , Porcinos , Transducción Genética
13.
Proc Natl Acad Sci U S A ; 101(22): 8313-8, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15159548

RESUMEN

Rab3 is a subfamily of the small GTP-binding protein Rab family and plays an important role in exocytosis. Several potential effectors of Rab3, including rabphilin3 and Rims (Rim1 and Rim2), have been isolated and characterized. Noc2 was identified originally in endocrine pancreas as a molecule homologous to rabphilin3, but its role in exocytosis is unclear. To clarify the physiological function of Noc2 directly, we have generated Noc2 knockout (Noc2(-/-)) mice. Glucose intolerance with impaired insulin secretion was induced in vivo by acute stress in Noc2(-/-) mice, but not in wild-type (Noc2(+/+)) mice. Ca(2+)-triggered insulin secretion from pancreatic isles of Noc2(-/-) mice was markedly impaired, but was completely restored by treatment with pertussis toxin, which inhibits inhibitory G protein Gi/o signaling. In addition, the inhibitory effect of clonidine, an alpha(2)-adrenoreceptor agonist, on insulin secretion was significantly greater in Noc2(-/-) islets than in Noc2(+/+) islets. Impaired Ca(2+)-triggered insulin secretion was rescued by adenovirus gene transfer of wild-type Noc2 but not by that of mutant Noc2, which does not bind to Rab3. Accordingly, Noc2 positively regulates insulin secretion from endocrine pancreas by inhibiting Gi/o signaling, and the interaction of Noc2 and Rab3 is required for the effect. Interestingly, we also found a marked accumulation of secretory granules in various exocrine cells of Noc2(-/-) mice, especially in exocrine pancreas with no amylase response to stimuli. Thus, Noc2, a critical effector of Rab3, is essential in normal regulation of exocytosis in both endocrine and exocrine cells.


Asunto(s)
Glándulas Exocrinas/metabolismo , Exocitosis/fisiología , Islotes Pancreáticos/metabolismo , Páncreas/citología , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Amilasas/metabolismo , Animales , Glucemia/metabolismo , Técnicas de Cultivo , Glándulas Exocrinas/citología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Noqueados , Páncreas/metabolismo , Toxina del Pertussis/metabolismo , Proteínas/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión al GTP rab3/metabolismo
14.
Circulation ; 107(5): 714-20, 2003 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-12578874

RESUMEN

BACKGROUND: Heart failure with preserved ejection fraction (HF-nlEF) is common in aged individuals with systolic hypertension and is frequently ascribed to diastolic dysfunction. We hypothesized that such patients also display combined ventricular-systolic and arterial stiffening that can exacerbate blood pressure lability and diastolic dysfunction under stress. METHODS AND RESULTS: Left ventricular pressure-volume relations were measured in patients with HF-nlEF (n=10) and contrasted with asymptomatic age-matched (n=9) and young (n=14) normotensives and age- and blood pressure-matched controls (n=25). End-systolic elastance (stiffness) was higher in patients with HF-nlEF (4.7+/-1.5 mm Hg/mL) than in controls (2.1+/-0.9 mm Hg/mL for normotensives and 3.3+/-1.0 mm Hg/mL for hypertensives; P<0.001). Effective arterial elastance was also higher (2.6+/-0.5 versus 1.9+/-0.5 mm Hg/mL) due to reduced total arterial compliance; the latter inversely correlated with end-systolic elastance (P=0.0001). Body size and stroke volumes were similar and could not explain differences in ventricular-arterial stiffening. HF-nlEF patients also displayed diastolic abnormalities, including higher left ventricular end-diastolic pressures (24.3+/-4.6 versus 12.9+/-5.5 mm Hg), caused by an upward-shifted diastolic pressure-volume curve. However, isovolumic relaxation and the early-to-late filling ratio were similar in age- and blood pressure-matched controls. Ventricular-arterial stiffening amplified stress-induced hypertension, which worsened diastolic function, and predicted higher cardiac energy costs to provide reserve output. CONCLUSION: Patients with HF-lnEF have systolic-ventricular and arterial stiffening beyond that associated with aging and/or hypertension. This may play an important pathophysiological role by exacerbating systemic load interaction with diastolic function, augmenting blood pressure lability, and elevating cardiac metabolic demand under stress.


Asunto(s)
Arterias/fisiopatología , Diástole , Insuficiencia Cardíaca/diagnóstico , Volumen Sistólico , Sístole , Disfunción Ventricular/diagnóstico , Adulto , Anciano , Demografía , Elasticidad , Femenino , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/fisiopatología , Pruebas de Función Cardíaca , Hemodinámica , Humanos , Masculino , Persona de Mediana Edad , Valores de Referencia , Disfunción Ventricular/complicaciones , Disfunción Ventricular/fisiopatología
15.
J Am Coll Cardiol ; 39(12): 2052-8, 2002 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12084608

RESUMEN

OBJECTIVES: This study sought to test a novel echocardiographic method based on contrast variability imaging (CVI), to quantify cardiac dyssynchrony and magnitude of resynchronization achieved by left ventricular (LV) and biventricular (BiV) pacing therapy. BACKGROUND: Left ventricular or BiV pacing is a promising new therapy for patients with heart failure and intraventricular conduction delay. However, precise quantitation of the extent of resynchronization achieved remains scant. METHODS: Ten patients treated with BiV or LV pacing therapy were studied. Echo-contrast was infused slowly, and gated images were acquired before and during contrast appearance. The temporally normalized variance derived from 30 to 50 sequential beats was determined at each pixel to yield the CVI image-displaying improved wall delineation. Systolic regional fractional area of radial sectors was calculated with active and temporarily suspended (AAI) pacing. All analyses were performed blinded to both patient and treatment. RESULTS: Pacing increased septal inward motion from -20.4 +/- 9.6% to -30.5 +/- 14.0%, whereas lateral wall motion occurred earlier with no net magnitude change. Both spatial and temporal dyssynchrony in the LV declined nearly 40% with LV or BiV pacing (p < or = 0.001), and this correlated with increasing ejection fraction (31% to 39%; p < 0.02; p < 0.004 for correlation with dyssynchrony). CONCLUSIONS: The new imaging and regional dyssynchrony analysis methods provide quantitative assessment of resynchronization analogous to that previously obtained only by tagged magnetic resonance imaging. This could provide a useful noninvasive method for both identifying candidates and following long-term therapy.


Asunto(s)
Estimulación Cardíaca Artificial , Cardiomiopatía Dilatada/diagnóstico por imagen , Ecocardiografía Doppler/métodos , Imagen de Acumulación Sanguínea de Compuerta/métodos , Corazón/diagnóstico por imagen , Humanos , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...