Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
BMC Endocr Disord ; 23(1): 133, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328745

RESUMEN

BACKGROUND: In the acute setting, PTH-independent hypercalcemia is typically treated with anti-resorptive agents such as zoledronic acid or denosumab. When these agents are no longer able to control hypercalcemia, several case reports have shown the utility of cinacalcet. However, it is not known if cinacalcet can be effective in patients naïve to anti-resorptive therapy or how cinacalcet ameliorates the hypercalcemia. CASE PRESENTATION: A 47-year-old male with a history of alcohol-induced cirrhosis was admitted for left cheek bleeding and swelling from an infiltrative squamous cell carcinoma of the oral cavity. On admission, he was found to have an elevated albumin-corrected serum calcium of 13.6 mg/dL, a serum phosphorus of 2.2 mg/dL and an intact PTH of 6 pg/mL (normal 18-90) with a PTHrP of 8.1 pmol/L (normal < 4.3), consistent with PTHrP-dependent hypercalcemia. Aggressive intravenous saline hydration and subcutaneous salmon calcitonin were initiated, but his serum calcium remained elevated. Given tooth extractions scheduled for the next day and possible irradiation to the jaw in the near future, alternatives to antiresorptive therapy were sought. Cinacalcet was initiated at 30 mg twice daily then increased to 60 mg twice daily the following day. The albumin-corrected serum calcium level decreased from 13.2 to 10.9 mg/dL within 48 h. The fractional excretion of calcium increased from 3.7 to 7.0%. CONCLUSIONS: This case demonstrates the utility of cinacalcet for the treatment of PTHrP-mediated hypercalcemia without prior anti-resorptive therapy via increased renal clearance of calcium.


Asunto(s)
Calcio , Hipercalcemia , Masculino , Humanos , Persona de Mediana Edad , Hipercalcemia/tratamiento farmacológico , Hipercalcemia/etiología , Cinacalcet/uso terapéutico , Proteína Relacionada con la Hormona Paratiroidea , Ácido Zoledrónico , Hormona Paratiroidea
2.
PLoS Genet ; 8(1): e1002449, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22253604

RESUMEN

The prevalence of type 2 diabetes in the United States is projected to double or triple by 2050. We reasoned that the genes that modulate insulin production might be new targets for diabetes therapeutics. Therefore, we developed an siRNA screening system to identify genes important for the activity of the insulin promoter in beta cells. We created a subclone of the MIN6 mouse pancreatic beta cell line that expresses destabilized GFP under the control of a 362 base pair fragment of the human insulin promoter and the mCherry red fluorescent protein under the control of the constitutively active rous sarcoma virus promoter. The ratio of the GFP to mCherry fluorescence of a cell indicates its insulin promoter activity. As G protein coupled receptors (GPCRs) have emerged as novel targets for diabetes therapies, we used this cell line to screen an siRNA library targeting all known mouse GPCRs. We identified several known GPCR regulators of insulin secretion as regulators of the insulin promoter. One of the top positive regulators was Gpr27, an orphan GPCR with no known role in beta cell function. We show that knockdown of Gpr27 reduces endogenous mouse insulin promoter activity and glucose stimulated insulin secretion. Furthermore, we show that Pdx1 is important for Gpr27's effect on the insulin promoter and insulin secretion. Finally, the over-expression of Gpr27 in 293T cells increases inositol phosphate levels, while knockdown of Gpr27 in MIN6 cells reduces inositol phosphate levels, suggesting this orphan GPCR might couple to Gq/11. In summary, we demonstrate a MIN6-based siRNA screening system that allows rapid identification of novel positive and negative regulators of the insulin promoter. Using this system, we identify Gpr27 as a positive regulator of insulin production.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Ratones , ARN Interferente Pequeño/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
3.
bioRxiv ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38746433

RESUMEN

Islet transplantation can cure type 1 diabetes, but peri-transplant beta cell death limits this procedure to those with low insulin requirements. Improving human beta cell survival or proliferation may make islet transplantation a possibility for more type 1 patients. To identify novel regulators of beta cell survival and proliferation, we conducted a pooled small hairpin RNA (shRNA) screen in primary human beta cells transplanted into immunocompromised mice. shRNAs targeting several cyclin dependent kinase inhibitors were enriched after transplant. Here, we focused on the Gi/o-coupled GPCR, serotonin 1F receptor ( HTR1F, 5-HT 1F ) which our screen identified as a negative regulator of beta cell numbers after transplant. In vitro , 5-HT 1F knockdown induced human beta cell proliferation but only when combined with harmine and exendin-4. In vivo , knockdown of 5-HT 1F reduced beta cell death during transplant. To demonstrate the feasibility of targeting 5-HT 1F in islet transplant, we identified and validated a small molecule 5-HT 1F antagonist. This antagonist increased glucose stimulated insulin secretion from primary human islets and cAMP accumulation in primary human beta cells. Finally, the 5-HT 1F antagonist improved glycemia in marginal mass, human islet transplants into immunocompromised mice. We identify 5-HT 1F as a novel druggable target to improve human beta cell survival in the setting of islet transplantation. One Sentence Summary: Serotonin 1F receptor (5-HT 1F ) negatively regulates insulin secretion and beta cell survival during transplant.

4.
bioRxiv ; 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38895340

RESUMEN

Imbalances in lipid storage and secretion lead to the accumulation of hepatocyte lipid droplets (LDs) (i.e., hepatic steatosis). Our understanding of the mechanisms that govern the channeling of hepatocyte neutral lipids towards cytosolic LDs or secreted lipoproteins remains incomplete. Here, we performed a series of CRISPR-Cas9 screens under different metabolic states to uncover mechanisms of hepatic neutral lipid flux. Clustering of chemical-genetic interactions identified CLIC-like chloride channel 1 (CLCC1) as a critical regulator of neutral lipid storage and secretion. Loss of CLCC1 resulted in the buildup of large LDs in hepatoma cells and knockout in mice caused liver steatosis. Remarkably, the LDs are in the lumen of the ER and exhibit properties of lipoproteins, indicating a profound shift in neutral lipid flux. Finally, remote homology searches identified a domain in CLCC1 that is homologous to yeast Brl1p and Brr6p, factors that promote the fusion of the inner and outer nuclear envelopes during nuclear pore complex assembly. Loss of CLCC1 lead to extensive nuclear membrane herniations, consistent with impaired nuclear pore complex assembly. Thus, we identify CLCC1 as the human Brl1p/Brr6p homolog and propose that CLCC1-mediated membrane remodeling promotes hepatic neutral lipid flux and nuclear pore complex assembly.

5.
PeerJ ; 11: e15077, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033725

RESUMEN

Understanding the interactions between SARS-CoV-2 and host cell machinery may reveal new targets to treat COVID-19. We focused on an interaction between the SARS-CoV-2 ORF3A accessory protein and the CLIC-like chloride channel-1 (CLCC1). We found that ORF3A partially co-localized with CLCC1 and that ORF3A and CLCC1 could be co-immunoprecipitated. Since CLCC1 plays a role in the unfolded protein response (UPR), we hypothesized that ORF3A may also play a role in the UPR. Indeed, ORF3A expression triggered a transcriptional UPR that was similar to knockdown of CLCC1. ORF3A expression in 293T cells induced cell death and this was rescued by the chemical chaperone taurodeoxycholic acid (TUDCA). Cells with CLCC1 knockdown were partially protected from ORF3A-mediated cell death. CLCC1 knockdown upregulated several of the homeostatic UPR targets induced by ORF3A expression, including HSPA6 and spliced XBP1, and these were not further upregulated by ORF3A. Our data suggest a model where CLCC1 silencing triggers a homeostatic UPR that prevents cell death due to ORF3A expression.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , COVID-19/genética , Canales de Cloruro/genética , Respuesta de Proteína Desplegada/genética , Muerte Celular
6.
ACS Sens ; 7(8): 2218-2224, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35951356

RESUMEN

Though the concentration of chloride has been measured in the cytoplasm and in secretory granules of live cells, it cannot be measured within the endoplasmic reticulum (ER) due to poor fluorescence of existing biosensors. We developed a fluorescent biosensor composed of a chloride-sensitive superfolder GFP and long Stokes-shifted mKate2 for simultaneous chloride and pH measurements that retained fluorescence in the ER lumen. Using this sensor, we showed that the chloride concentration in the ER is significantly lower than that in the cytosol. This improved biosensor enables dynamic measurement of chloride in the ER and may be useful in other environments where protein folding is challenging.


Asunto(s)
Técnicas Biosensibles , Cloruros , Retículo Endoplásmico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Pliegue de Proteína
7.
Front Genet ; 13: 804924, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35391798

RESUMEN

Background: A CLCC1 c. 75C > A (p.D25E) mutation has been associated with autosomal recessive pigmentosa in patients in and from Pakistan. CLCC1 is ubiquitously expressed, and knockout models of this gene in zebrafish and mice are lethal in the embryonic period, suggesting that possible retinitis pigmentosa mutations in this gene might be limited to those leaving partial activity. In agreement with this hypothesis, the mutation is the only CLCC1 mutation associated with retinitis pigmentosa to date, and all identified patients with this mutation share a common SNP haplotype surrounding the mutation, suggesting a common founder. Methods: SNPs were genotyped by a combination of WGS and Sanger sequencing. The original founder haplotype, and recombination pathways were delineated by examination to minimize recombination events. Mutation age was estimated by four methods including an explicit solution, an iterative approach, a Bayesian approach and an approach based solely on ancestral segment lengths using high density SNP data. Results: All members of each of the nine families studied shared a single autozygous SNP haplotype for the CLCC1 region ranging from approximately 1-3.5 Mb in size. The haplotypes shared by the families could be derived from a single putative ancestral haplotype with at most two recombination events. Based on the haplotype and Gamma analysis, the estimated age of the founding mutation varied from 79 to 196 generations, or approximately 2,000-5,000 years, depending on the markers used in the estimate. The DMLE (Bayesian) estimates ranged from 2,160 generations assuming a population growth rate of 0-309 generations assuming a population growth rate of 2% with broad 95% confidence intervals. Conclusion: These results provide insight into the origin of the CLCC1 mutation in the Pakistan population. This mutation is estimated to have occurred 2000-5,000 years ago and has been transmitted to affected families of Pakistani origin in geographically dispersed locations around the world. This is the only mutation in CLCC1 identified to date, suggesting that the CLCC1 gene is under a high degree of constraint, probably imposed by functional requirements for this gene during embryonic development.

8.
Cell Metab ; 33(7): 1322-1341.e13, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34019840

RESUMEN

Mitochondria control eukaryotic cell fate by producing the energy needed to support life and the signals required to execute programed cell death. The biochemical milieu is known to affect mitochondrial function and contribute to the dysfunctional mitochondrial phenotypes implicated in cancer and the morbidities of aging. However, the physical characteristics of the extracellular matrix are also altered in cancerous and aging tissues. Here, we demonstrate that cells sense the physical properties of the extracellular matrix and activate a mitochondrial stress response that adaptively tunes mitochondrial function via solute carrier family 9 member A1-dependent ion exchange and heat shock factor 1-dependent transcription. Overall, our data indicate that adhesion-mediated mechanosignaling may play an unappreciated role in the altered mitochondrial functions observed in aging and cancer.


Asunto(s)
Adhesión Celular/fisiología , Mecanotransducción Celular/fisiología , Dinámicas Mitocondriales/fisiología , Adulto , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Respiración de la Célula , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Células HEK293 , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hiperglucemia/fisiopatología , Integrinas/fisiología , Intercambio Iónico , Ratones , Microscopía Confocal , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/fisiología , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Intercambiador 1 de Sodio-Hidrógeno/fisiología , Imagen de Lapso de Tiempo
9.
Sci Rep ; 10(1): 5629, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32221326

RESUMEN

Gpr27 is a highly conserved, orphan G protein coupled receptor (GPCR) previously implicated in pancreatic beta cell insulin transcription and glucose-stimulated insulin secretion in vitro. Here, we characterize a whole-body mouse knockout of Gpr27. Gpr27 knockout mice were born at expected Mendelian ratios and exhibited no gross abnormalities. Insulin and Pdx1 mRNA in Gpr27 knockout islets were reduced by 30%, but this did not translate to a reduction in islet insulin content or beta cell mass. Gpr27 knockout mice exhibited slightly worsened glucose tolerance with lower plasma insulin levels while maintaining similar insulin tolerance. Unexpectedly, Gpr27 deletion reduced expression of Eif4e3, a neighboring gene, likely by deleting transcription start sites on the anti-sense strand of the Gpr27 coding exon. Our data confirm that loss of Gpr27 reduces insulin mRNA in vivo but has only minor effects on glucose tolerance.


Asunto(s)
Diabetes Mellitus/metabolismo , Insulina/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Glucosa/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Noqueados
10.
Islets ; 12(3): 59-69, 2020 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-32579048

RESUMEN

Virus-mediated gene knockdown in intact pancreatic islets is technically challenging due to poor infection of the center of the islet. Because the cells that do not have knockdown have normal insulin secretion, measuring changes in insulin secretion after gene knockdown is challenging. We describe a method to monitor insulin secretion from only the beta cells with knockdown of a gene of interest in intact islets using a single lentivirus containing a guide RNA, a luciferase insulin secretion reporter and a dCas9-KRAB cassette. This method allows rapid and inexpensive monitoring of insulin secretion from only those beta cells with knockdown, circumventing the problem of incomplete islet infection.


Asunto(s)
Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Edición Génica , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Secreciones Corporales , Edición Génica/métodos , Técnicas de Silenciamiento del Gen , Ratones , Interferencia de ARN
11.
Hum Gene Ther ; 19(1): 17-26, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18211225

RESUMEN

RNA interference (RNAi) is a sequence-specific gene-silencing phenomenon mediated by double-stranded RNA. RNAi is made possible through the activity of an evolutionarily conserved RNA-protein machinery that is utilized by microRNAs (miRNAs), endogenously produced 21- to 23-nucleotide noncoding RNAs. As RNA-based therapeutics come of age, understanding the miRNA pathway and the mechanism of posttranscriptional gene silencing is paramount. Studies have pointed to a critical role for miRNAs in human development and disease and revealed an unanticipated complexity in the mechanisms of regulation of gene expression. This review discusses advances in the biogenesis of miRNAs, their modes of action, and implications for RNA-based treatments.


Asunto(s)
MicroARNs/metabolismo , Interferencia de ARN , Humanos , Modelos Biológicos , Transducción de Señal
12.
Cell Stem Cell ; 22(3): 369-383.e8, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29499153

RESUMEN

A permissive chromatin environment coupled to hypertranscription drives the rapid proliferation of embryonic stem cells (ESCs) and peri-implantation embryos. We carried out a genome-wide screen to systematically dissect the regulation of the euchromatic state of ESCs. The results revealed that cellular growth pathways, most prominently translation, perpetuate the euchromatic state and hypertranscription of ESCs. Acute inhibition of translation rapidly depletes euchromatic marks in mouse ESCs and blastocysts, concurrent with delocalization of RNA polymerase II and reduction in nascent transcription. Translation inhibition promotes rewiring of chromatin accessibility, which decreases at a subset of active developmental enhancers and increases at histone genes and transposable elements. Proteome-scale analyses revealed that several euchromatin regulators are unstable proteins and continuously depend on a high translational output. We propose that this mechanistic interdependence of euchromatin, transcription, and translation sets the pace of proliferation at peri-implantation and may be employed by other stem/progenitor cells.


Asunto(s)
Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Biosíntesis de Proteínas , Transcripción Genética , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Elementos Transponibles de ADN/genética , Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos/genética , Eucromatina/metabolismo , Femenino , Genoma , Código de Histonas , Masculino , Ratones , Modelos Biológicos , Proteínas Nucleares/metabolismo , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Serina-Treonina Quinasas TOR/metabolismo
13.
Endocrinology ; 159(9): 3245-3256, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30052866

RESUMEN

Mitochondria are dynamic organelles that undergo frequent fission and fusion events. Mitochondrial fission is required for ATP production, the tricarboxylic acid cycle, and processes beyond metabolism in a cell-type specific manner. Ex vivo and cell line studies have demonstrated that Drp1, a central regulator of mitochondrial fission, is required for glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells. Herein, we set out to interrogate the role of Drp1 in ß-cell insulin secretion in vivo. We generated ß-cell-specific Drp1 knockout (KO) mice (Drp1ß-KO) by crossing a conditional allele of Drp1 to Ins1cre mice, in which Cre recombinase replaces the coding region of the Ins1 gene. Drp1ß-KO mice were glucose intolerant due to impaired GSIS but did not progress to fasting hyperglycemia as adults. Despite markedly abnormal mitochondrial morphology, Drp1ß-KO islets exhibited normal oxygen consumption rates and an unchanged glucose threshold for intracellular calcium mobilization. Instead, the most profound consequences of ß-cell Drp1 deletion were impaired second-phase insulin secretion and impaired glucose-stimulated amplification of insulin secretion. Our data establish Drp1 as an important regulator of insulin secretion in vivo and demonstrate a role for Drp1 in metabolic amplification and calcium handling without affecting oxygen consumption.


Asunto(s)
Dinaminas/genética , Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Consumo de Oxígeno/genética , Animales , Calcio/metabolismo , Ayuno/metabolismo , Intolerancia a la Glucosa/genética , Hiperglucemia/genética , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/patología , Dinámicas Mitocondriales
14.
Endocrinology ; 159(9): 3321-3330, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30059978

RESUMEN

Insulin production by the pancreatic ß cell is critical for the glucose homeostasis of the whole organism. Although the transcription factors required for insulin production are known, the upstream pathways that control insulin production are less clear. To further elucidate this regulatory network, we created a genetic interaction map of insulin production by performing ∼20,000 pairwise RNA interference knockdowns of insulin promoter regulators. Our map correctly predicted known physical complexes in the electron transport chain and a role for Spry2 in the unfolded protein response. To further validate our map, we used it to predict the function of an unannotated gene encoding a 37-kDa protein with no identifiable domains we have termed mitochondrial fission factor interactor (Mfi). We have shown that Mfi is a binding partner of the mitochondrial fission factor and that Mfi inhibits dynamin-like protein 1 recruitment to mitochondria. Our data provide a resource to understand the regulatory network of insulin promoter activity.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Mitocondrias/metabolismo , Dinámicas Mitocondriales/genética , Animales , Línea Celular , Dinaminas , GTP Fosfohidrolasas , Redes Reguladoras de Genes , Humanos , Insulina/genética , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos , Proteínas Mitocondriales/metabolismo , Regiones Promotoras Genéticas/genética , Respuesta de Proteína Desplegada
15.
Diabetes ; 66(6): 1703-1712, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28246293

RESUMEN

Insulin production by the pancreatic ß-cell is required for normal glucose homeostasis. While key transcription factors that bind to the insulin promoter are known, relatively little is known about the upstream regulators of insulin transcription. Using a whole-genome RNA interference screen, we uncovered 26 novel regulators of insulin transcription that regulate diverse processes including oxidative phosphorylation, vesicle traffic, and the unfolded protein response (UPR). We focused on Spry2-a gene implicated in human type 2 diabetes by genome-wide association studies but without a clear connection to glucose homeostasis. We showed that Spry2 is a novel UPR target and its upregulation is dependent on PERK. Knockdown of Spry2 resulted in reduced expression of Serca2, reduced endoplasmic reticulum calcium levels, and induction of the UPR. Spry2 deletion in the adult mouse ß-cell caused hyperglycemia and hypoinsulinemia. Our study greatly expands the compendium of insulin promoter regulators and demonstrates a novel ß-cell link between Spry2 and human diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Regulación de la Expresión Génica/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Respuesta de Proteína Desplegada/genética , Animales , Anexina A5/metabolismo , Western Blotting , Calcio/metabolismo , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , eIF-2 Quinasa/metabolismo
16.
J Clin Invest ; 124(9): 4093-101, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25133424

RESUMEN

Endocrine cell proliferation fluctuates dramatically in response to signals that communicate hormone demand. The genetic alterations that override these controls in endocrine tumors often are not associated with oncogenes common to other tumor types, suggesting that unique pathways govern endocrine proliferation. Within the pancreas, for example, activating mutations of the prototypical oncogene KRAS drive proliferation in all pancreatic ductal adenocarcimomas but are never found in pancreatic endocrine tumors. Therefore, we asked how cellular context impacts K-RAS signaling. We found that K-RAS paradoxically suppressed, rather than promoted, growth in pancreatic endocrine cells. Inhibition of proliferation by K-RAS depended on antiproliferative RAS effector RASSF1A and blockade of the RAS-activated proproliferative RAF/MAPK pathway by tumor suppressor menin. Consistent with this model, a glucagon-like peptide 1 (GLP1) agonist, which stimulates ERK1/2 phosphorylation, did not affect endocrine cell proliferation by itself, but synergistically enhanced proliferation when combined with a menin inhibitor. In contrast, inhibition of MAPK signaling created a synthetic lethal interaction in the setting of menin loss. These insights suggest potential strategies both for regenerating pancreatic ß cells for people with diabetes and for targeting menin-sensitive endocrine tumors.


Asunto(s)
Islotes Pancreáticos/citología , Proteínas Proto-Oncogénicas/fisiología , Proteínas ras/fisiología , Adulto , Animales , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Proteínas Supresoras de Tumor/fisiología
17.
Mol Endocrinol ; 26(10): 1783-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22915829

RESUMEN

The pancreatic ß-cell is critical for the maintenance of glycemic control. Knowing the compendium of genes expressed in ß-cells will further our understanding of this critical cell type and may allow the identification of future antidiabetes drug targets. Here, we report the use of next-generation sequencing to obtain nearly 1 billion reads from the polyadenylated RNA of islets and purified ß-cells from mice. These data reveal novel examples of ß-cell-specific splicing events, promoter usage, and over 1000 long intergenic noncoding RNA expressed in mouse ß-cells. Many of these long intergenic noncoding RNA are ß-cell specific, and we hypothesize that this large set of novel RNA may play important roles in ß-cell function. Our data demonstrate unique features of the ß-cell transcriptome.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Células Secretoras de Insulina/metabolismo , Transcriptoma , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos , Regiones Promotoras Genéticas , Empalme del ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA