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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 168(1-2): 73-85.e11, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-27916274

RESUMEN

The recent discovery that genetically modified α cells can regenerate and convert into ß-like cells in vivo holds great promise for diabetes research. However, to eventually translate these findings to human, it is crucial to discover compounds with similar activities. Herein, we report the identification of GABA as an inducer of α-to-ß-like cell conversion in vivo. This conversion induces α cell replacement mechanisms through the mobilization of duct-lining precursor cells that adopt an α cell identity prior to being converted into ß-like cells, solely upon sustained GABA exposure. Importantly, these neo-generated ß-like cells are functional and can repeatedly reverse chemically induced diabetes in vivo. Similarly, the treatment of transplanted human islets with GABA results in a loss of α cells and a concomitant increase in ß-like cell counts, suggestive of α-to-ß-like cell conversion processes also in humans. This newly discovered GABA-induced α cell-mediated ß-like cell neogenesis could therefore represent an unprecedented hope toward improved therapies for diabetes.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Ácido gamma-Aminobutírico/administración & dosificación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Células Secretoras de Glucagón/efectos de los fármacos , Humanos , Islotes Pancreáticos/citología , Masculino , Ratones , Proteínas del Tejido Nervioso , Ratas , Ratas Wistar , Ácido gamma-Aminobutírico/farmacología
2.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-27916275

RESUMEN

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Asunto(s)
Artemisininas/farmacología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Modelos Animales de Enfermedad , Receptores de GABA-A/metabolismo , Transducción de Señal , Animales , Arteméter , Artemisininas/administración & dosificación , Proteínas Portadoras/metabolismo , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus Tipo 1/patología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Estabilidad Proteica/efectos de los fármacos , Ratas , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Pez Cebra , Ácido gamma-Aminobutírico/metabolismo
3.
Cell ; 138(3): 449-62, 2009 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-19665969

RESUMEN

We have previously reported that the loss of Arx and/or Pax4 gene activity leads to a shift in the fate of the different endocrine cell subtypes in the mouse pancreas, without affecting the total endocrine cell numbers. Here, we conditionally and ectopically express Pax4 using different cell-specific promoters and demonstrate that Pax4 forces endocrine precursor cells, as well as mature alpha cells, to adopt a beta cell destiny. This results in a glucagon deficiency that provokes a compensatory and continuous glucagon+ cell neogenesis requiring the re-expression of the proendocrine gene Ngn3. However, the newly formed alpha cells fail to correct the hypoglucagonemia since they subsequently acquire a beta cell phenotype upon Pax4 ectopic expression. Notably, this cycle of neogenesis and redifferentiation caused by ectopic expression of Pax4 in alpha cells is capable of restoring a functional beta cell mass and curing diabetes in animals that have been chemically depleted of beta cells.


Asunto(s)
Diferenciación Celular , Células Secretoras de Glucagón/citología , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Factores de Transcripción Paired Box/metabolismo , Páncreas/citología , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Glucagón/deficiencia , Islotes Pancreáticos/citología , Ratones , Proteínas del Tejido Nervioso/metabolismo , Páncreas/crecimiento & desarrollo
4.
Hum Mol Genet ; 28(24): 4089-4102, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31691806

RESUMEN

A disproportional large number of neurodevelopmental disorders (NDDs) is caused by variants in genes encoding transcription factors and chromatin modifiers. However, the functional interactions between the corresponding proteins are only partly known. Here, we show that KDM5C, encoding a H3K4 demethylase, is at the intersection of transcriptional axes under the control of three regulatory proteins ARX, ZNF711 and PHF8. Interestingly, mutations in all four genes (KDM5C, ARX, ZNF711 and PHF8) are associated with X-linked NDDs comprising intellectual disability as a core feature. in vitro analysis of the KDM5C promoter revealed that ARX and ZNF711 function as antagonist transcription factors that activate KDM5C expression and compete for the recruitment of PHF8. Functional analysis of mutations in these genes showed a correlation between phenotype severity and the reduction in KDM5C transcriptional activity. The KDM5C decrease was associated with a lack of repression of downstream target genes Scn2a, Syn1 and Bdnf in the embryonic brain of Arx-null mice. Aiming to correct the faulty expression of KDM5C, we studied the effect of the FDA-approved histone deacetylase inhibitor suberanilohydroxamic acid (SAHA). In Arx-KO murine ES-derived neurons, SAHA was able to rescue KDM5C depletion, recover H3K4me3 signalling and improve neuronal differentiation. Indeed, in ARX/alr-1-deficient Caenorhabditis elegans animals, SAHA was shown to counteract the defective KDM5C/rbr-2-H3K4me3 signalling, recover abnormal behavioural phenotype and ameliorate neuronal maturation. Overall, our studies indicate that KDM5C is a conserved and druggable effector molecule across a number of NDDs for whom the use of SAHA may be considered a potential therapeutic strategy.


Asunto(s)
Histona Demetilasas/metabolismo , Trastornos del Neurodesarrollo/metabolismo , Animales , Caenorhabditis elegans , Línea Celular , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Inhibidores de Histona Desacetilasas/farmacología , Histona Demetilasas/genética , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Trastornos del Neurodesarrollo/genética , Neuronas/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/metabolismo , Vorinostat/farmacología
5.
Diabetes Obes Metab ; 20 Suppl 2: 3-10, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30230184

RESUMEN

Ghrelin is a gastric peptide with anabolic functions. It acutely stimulates growth hormone (GH) secretion from the anterior pituitary glands and modulates hypothalamic circuits that control food intake and energy expenditure. Besides its central activity, ghrelin is also involved in the regulation of pancreatic development and physiology. Particularly, several studies highlighted the ability of ghrelin to sustain ß-cell viability and proliferation. Furthermore, ghrelin seems to exert inhibitory effects on pancreatic acinar and endocrine secretory functions. Due to its pleiotropic activity on energy metabolism, ghrelin has become a topic of great interest for experimental research focused on type II diabetes and obesity. The aim of this review is to illustrate the complex and not fully understood interplay between ghrelin, pancreas and glucose homeostasis.


Asunto(s)
Ghrelina/fisiología , Páncreas/crecimiento & desarrollo , Animales , Glucemia/metabolismo , Diabetes Mellitus/etiología , Ghrelina/genética , Homeostasis/fisiología , Humanos , Hipotálamo/fisiología , Ratones , Páncreas/fisiología , Receptores de Ghrelina/fisiología
6.
Semin Cell Dev Biol ; 44: 107-14, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26319183

RESUMEN

The embryonic development of the pancreas is orchestrated by a complex and coordinated transcription factor network. Neurogenin3 (Neurog3) initiates the endocrine program by activating the expression of additional transcription factors driving survival, proliferation, maturation and lineage allocation of endocrine precursors. Among the direct targets of Neurog3, Pax4 appears as one of the key regulators of ß-cell specification. Indeed, mice lacking Pax4 die a few days postpartum, as they develop severe hyperglycemia due to the absence of mature pancreatic ß-cells. Pax4 also directly regulates the expression of Arx, a gene that plays a crucial role in α-cell specification. Comparative analysis of Pax4 and Arx mutants, as well as Arx/Pax4 double mutants, showed that islet subtype destiny is mainly directed by cross-repression of the Pax4 and Arx factors. Importantly, the ectopic expression of Pax4 in α-cells was found sufficient to induce their neogenesis and conversion into ß-like cells, not only during development but also in adult rodents. Therefore, differentiated endocrine α-cells can be considered as a putative source for insulin-producing ß-like cells. These findings have clearly widened our understanding regarding pancreatic development, but they also open new research avenues in the context of diabetes research.


Asunto(s)
Proteínas de Homeodominio/fisiología , Factores de Transcripción Paired Box/fisiología , Animales , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Páncreas/embriología , Páncreas/metabolismo , Páncreas/fisiología
7.
Am J Hum Genet ; 92(1): 114-25, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23246292

RESUMEN

Intellectual disability (ID) and epilepsy often occur together and have a dramatic impact on the development and quality of life of the affected children. Polyalanine (polyA)-expansion-encoding mutations of aristaless-related homeobox (ARX) cause a spectrum of X-linked ID (XLID) diseases and chronic epilepsy, including infantile spasms. We show that lysine-specific demethylase 5C (KDM5C), a gene known to be mutated in XLID-affected children and involved in chromatin remodeling, is directly regulated by ARX through the binding in a conserved noncoding element. We have studied altered ARX carrying various polyA elongations in individuals with XLID and/or epilepsy. The changes in polyA repeats cause hypomorphic ARX alterations, which exhibit a decreased trans-activity and reduced, but not abolished, binding to the KDM5C regulatory region. The altered functioning of the mutants tested is likely to correlate with the severity of XLID and/or epilepsy. By quantitative RT-PCR, we observed a dramatic Kdm5c mRNA downregulation in murine Arx-knockout embryonic and neural stem cells. Such Kdm5c mRNA diminution led to a severe decrease in the KDM5C content during in vitro neuronal differentiation, which inversely correlated with an increase in H3K4me3 signal. We established that ARX polyA alterations damage the regulation of KDM5C expression, and we propose a potential ARX-dependent path acting via chromatin remodeling.


Asunto(s)
Epilepsia/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Oxidorreductasas N-Desmetilantes/genética , Factores de Transcripción/genética , Animales , Niño , Expansión de las Repeticiones de ADN , Histona Demetilasas , Humanos , Ratones , Ratones Noqueados , Péptidos/genética
8.
PLoS Genet ; 9(10): e1003934, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204325

RESUMEN

Recently, it was demonstrated that pancreatic new-born glucagon-producing cells can regenerate and convert into insulin-producing ß-like cells through the ectopic expression of a single gene, Pax4. Here, combining conditional loss-of-function and lineage tracing approaches, we show that the selective inhibition of the Arx gene in α-cells is sufficient to promote the conversion of adult α-cells into ß-like cells at any age. Interestingly, this conversion induces the continuous mobilization of duct-lining precursor cells to adopt an endocrine cell fate, the glucagon(+) cells thereby generated being subsequently converted into ß-like cells upon Arx inhibition. Of interest, through the generation and analysis of Arx and Pax4 conditional double-mutants, we provide evidence that Pax4 is dispensable for these regeneration processes, indicating that Arx represents the main trigger of α-cell-mediated ß-like cell neogenesis. Importantly, the loss of Arx in α-cells is sufficient to regenerate a functional ß-cell mass and thereby reverse diabetes following toxin-induced ß-cell depletion. Our data therefore suggest that strategies aiming at inhibiting the expression of Arx, or its molecular targets/co-factors, may pave new avenues for the treatment of diabetes.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus Tipo 1/genética , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/metabolismo , Factores de Transcripción/genética , Animales , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucagón/genética , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/biosíntesis , Humanos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones Transgénicos , Factores de Transcripción Paired Box/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/biosíntesis
9.
Med Sci (Paris) ; 29(8-9): 749-55, 2013.
Artículo en Francés | MEDLINE | ID: mdl-24005630

RESUMEN

Type 1 diabetes (T1DM) is a common metabolic disorder affecting an ever-increasing number of patients worldwide. T1DM is caused by the selective destruction of pancreatic insulin-producing ß-cells by the immune system. Such loss results in chronic hyperglycemia and could induce a number of cardio-vascular complications. Despite the classical insulin-based therapy, compared to healthy people, patients with T1DM display a shortened life expectancy due to the treatment's inability to strictly regulate glycemic levels. An alternative therapy involves pancreatic islet transplantation but the shortage of donors and the required immuno-suppressive treatments limit the widespread use of this approach. Therefore, the search of new approaches to generate functional ß-cells is of growing interest. In this review, we describe several novel strategies aiming at the conversion of diverse pancreatic cells into ß-cells, such as acinar, ductal, and endocrine cells. Clearly, such promising results could open new research avenues in the context of type 1 diabetes research.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/citología , Páncreas/citología , Células Acinares/citología , Diabetes Mellitus Tipo 1/cirugía , Humanos , Células Secretoras de Insulina/fisiología , Trasplante de Islotes Pancreáticos , Conductos Pancreáticos/citología , Regeneración , Donantes de Tejidos/provisión & distribución
10.
Cells ; 12(4)2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36831232

RESUMEN

The pancreas is an abdominal gland that serves 2 vital purposes: assist food processing by secreting digestive enzymes and regulate blood glucose levels by releasing endocrine hormones. During embryonic development, this gland originates from epithelial buds located on opposite sites of the foregut endoderm. Pancreatic cell specification and maturation are coordinated by a complex interplay of extrinsic and intrinsic signaling events. In the recent years, the canonical Wnt/ß-catenin pathway has emerged as an important player of pancreas organogenesis, regulating pancreatic epithelium specification, compartmentalization and expansion. Importantly, it has been suggested to regulate proliferation, survival and function of adult pancreatic cells, including insulin-secreting ß-cells. This review summarizes recent work on the role of Wnt/ß-catenin signaling in pancreas biology from early development to adulthood, emphasizing on its relevance for the development of new therapies for pancreatic diseases.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Embarazo , Femenino , Humanos , beta Catenina/metabolismo , Páncreas/metabolismo , Organogénesis , Desarrollo Embrionario
11.
Diabetes ; 72(8): 1112-1126, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37216637

RESUMEN

The loss of pancreatic ß-cell identity has emerged as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell-cycle regulator and transcription factor E2F1 in the maintenance of ß-cell identity, insulin secretion, and glucose homeostasis. We show that the ß-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, altered endocrine cell mass, downregulation of many ß-cell genes, and concomitant increase of non-ß-cell markers. Mechanistically, epigenomic profiling of the promoters of these non-ß-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic, and epigenomic signatures are associated with these ß-cell dysfunctions, with E2F1 directly regulating several ß-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of ß-cell identity genes. Our data suggest that E2F1 is critical for maintaining ß-cell identity and function through sustained control of ß-cell and non-ß-cell transcriptional programs. ARTICLE HIGHLIGHTS: ß-Cell-specific E2f1 deficiency in mice impairs glucose tolerance. Loss of E2f1 function alters the ratio of α- to ß-cells but does not trigger ß-cell conversion into α-cells. Pharmacological inhibition of E2F activity inhibits glucose-stimulated insulin secretion and alters ß- and α-cell gene expression in human islets. E2F1 maintains ß-cell function and identity through control of transcriptomic and epigenetic programs.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Humanos , Ratones , Cromatina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Histonas/metabolismo , Homeostasis/genética , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones Noqueados
12.
Semin Cell Dev Biol ; 21(8): 838-44, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20688184

RESUMEN

The pancreas is composed of two main compartments consisting of endocrine and exocrine tissues. The majority of the organ is exocrine and responsible for the synthesis of digestive enzymes and for their transport via an intricate ductal system into the duodenum. The endocrine tissue represents less than 2% of the organ and is organized into functional units called islets of Langerhans, comprising alpha-, beta-, delta-, epsilon- and PP-cells, producing the hormones glucagon, insulin, somatostatin, ghrelin and pancreatic polypeptide (PP), respectively. Insulin-producing beta-cells play a central role in the control of the glucose homeostasis. Accordingly, absolute or relative deficiency in beta-cells may ultimately lead to type 1 and/or type 2 diabetes, respectively. One major goal of diabetes research is therefore to understand the molecular mechanisms controlling the development of beta-cells during pancreas morphogenesis, but also those underlying the regeneration of adult injured pancreas, and assess their significance for future cell-based therapy. In this review, we will therefore present new insights into beta-cell development with focus on beta-cell regeneration.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Animales , Proliferación Celular , Humanos , Ratones , Páncreas/citología , Páncreas/embriología
13.
Transgenic Res ; 21(6): 1215-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22717987

RESUMEN

Pax4 belongs to the paired-box family of transcription factors. The analysis of loss- and gain-of-function mutant animals revealed that this factor plays a crucial role in the endocrine pancreas. Indeed, Pax4 is required for the genesis of insulin-producing beta-cells. Remarkably, the sole misexpression of Pax4 in glucagon-expressing cells is able to induce their regeneration, endow these with beta-cell features, and thereby counter chemically induced diabetes. However, the function of Pax4 in adult endocrine cells remains unclear. Herein, we report the generation of Pax4 conditional knockout mice that will allow the analysis of Pax4 function in mature beta-cells, as well as in the adult central nervous system.


Asunto(s)
Proteínas de Homeodominio/fisiología , Células Secretoras de Insulina/metabolismo , Integrasas/metabolismo , Factores de Transcripción Paired Box/fisiología , Hormonas Pancreáticas/metabolismo , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Técnicas para Inmunoenzimas , Células Secretoras de Insulina/citología , Ratones , Ratones Noqueados
14.
Front Endocrinol (Lausanne) ; 13: 861922, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573999

RESUMEN

Type 1 diabetes results from the autoimmune-mediated loss of insulin-producing beta-cells. Accordingly, important research efforts aim at regenerating these lost beta-cells by converting pre-existing endogenous cells. Following up on previous results demonstrating the conversion of pancreatic somatostatin delta-cells into beta-like cells upon Pax4 misexpression and acknowledging that somatostatin-expressing cells are highly represented in the gastrointestinal tract, one could wonder whether this Pax4-mediated conversion could also occur in the GI tract. We made use of transgenic mice misexpressing Pax4 in somatostatin cells (SSTCrePOE) to evaluate a putative Pax4-mediated D-to-beta-like cell conversion. Additionally, we implemented an ex vivo approach based on mice-derived gut organoids to assess the functionality of these neo-generated beta-like cells. Our results outlined the presence of insulin+ cells expressing several beta-cell markers in gastrointestinal tissues of SSTCrePOE animals. Further, using lineage tracing, we established that these cells arose from D cells. Lastly, functional tests on mice-derived gut organoids established the ability of neo-generated beta-like cells to release insulin upon stimulation. From this study, we conclude that the misexpression of Pax4 in D cells appears sufficient to convert these into functional beta-like cells, thus opening new research avenues in the context of diabetes research.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción Paired Box/metabolismo , Células Secretoras de Somatostatina , Animales , Proteínas de Homeodominio/genética , Insulina , Ratones , Factores de Transcripción Paired Box/genética , Somatostatina/genética
15.
BMC Dev Biol ; 11: 52, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21880149

RESUMEN

BACKGROUND: Nkx2.2 and Arx represent key transcription factors implicated in the specification of islet cell subtypes during pancreas development. Mice deficient for Arx do not develop any alpha-cells whereas beta- and delta-cells are found in considerably higher numbers. In Nkx2.2 mutant animals, alpha- and beta-cell development is severely impaired whereas a ghrelin-expressing cell population is found augmented.Notably, Arx transcription is clearly enhanced in Nkx2.2-deficient pancreata. Hence in order to precise the functional link between both factors we performed a comparative analysis of Nkx2.2/Arx single- and double-mutants but also of Pax6-deficient animals. RESULTS: We show that most of the ghrelin+ cells emerging in pancreata of Nkx2.2- and Pax6-deficient mice, express the alpha-cell specifier Arx, but also additional beta-cell related genes. In Nkx2.2-deficient mice, Arx directly co-localizes with iAPP, PC1/3 and Pdx1 suggesting an Nkx2.2-dependent control of Arx in committed beta-cells. The combined loss of Nkx2.2 and Arx likewise results in the formation of a hyperplastic ghrelin+ cell population at the expense of mature alpha- and beta-cells. Surprisingly, such Nkx2.2-/-Arx- ghrelin+ cells also express the somatostatin hormone. CONCLUSIONS: Our data indicate that Nkx2.2 acts by reinforcing the transcriptional networks initiated by Pax4 and Arx in early committed beta- and alpha-cell, respectively. Our analysis also suggests that one of the coupled functions of Nkx2.2 and Pax4 is to counteract Arx gene activity in early committed beta-cells.


Asunto(s)
Linaje de la Célula , Ghrelina/biosíntesis , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/biosíntesis , Células Secretoras de Insulina/metabolismo , Somatostatina/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Ghrelina/genética , Células Secretoras de Glucagón/citología , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Factor de Transcripción PAX6 , Factores del Dominio POU/biosíntesis , Factores de Transcripción Paired Box/biosíntesis , Factores de Transcripción Paired Box/deficiencia , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/biosíntesis , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Somatostatina/genética , Transactivadores/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas de Pez Cebra
16.
NPJ Regen Med ; 6(1): 7, 2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33580013

RESUMEN

The role of DNA methylation in ß-cell neogenesis is poorly understood. We report that during the process of induced cell reprogramming, methylation content of the Ngn3 and Sox11 genes are diminished. These findings emphasise DNA methylation is a barrier in ß-cell regeneration in adulthood, a well described pathophysiological phenomenon of major significance in explaining ß-cell deficiency in diabetes in the adult pancreas.

17.
Cells ; 10(11)2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34831029

RESUMEN

Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on ß-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of ß-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/prevención & control , Factores de Transcripción/deficiencia , Células Acinares/citología , Células Acinares/metabolismo , Amilasas/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ghrelina/metabolismo , Proteínas de Homeodominio/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/genética , Integrasas/metabolismo , Ratones Transgénicos , Mutación/genética , Páncreas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Dev Biol ; 334(1): 59-71, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19627984

RESUMEN

The homeobox-containing gene Arx is expressed during ventral telencephalon development and required for correct GABAergic interneuron tangential migration from the ganglionic eminences to the olfactory bulbs, cerebral cortex and striatum. Its human ortholog is associated with a variety of neurological clinical manifestations whose symptoms are compatible with the loss of cortical interneurons and altered basal ganglia-related activities. Herein, we report the identification of a number of genes whose expression is consistently altered in Arx mutant ganglionic eminences. Our analyses revealed a striking ectopic expression in the ganglionic eminences of several of these genes normally at most marginally expressed in the ventral telencephalon. Among them, Ebf3 was functionally analyzed. Thus, its ectopic expression in ventral telencephalon was found to prevent neuronal tangential migration. Further, we showed that Arx is sufficient to repress Ebf3 endogenous expression and that its silencing in Arx mutant tissues partially rescues tangential cell movement. Together, these data provide new insights into the molecular pathways regulated by Arx during telencephalon development.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas Represoras/genética , Telencéfalo/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Animales , Ganglios Basales/metabolismo , Movimiento Celular , Regulación hacia Abajo , Embrión de Mamíferos/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo
19.
J Clin Invest ; 117(4): 961-70, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17404619

RESUMEN

Aristaless-related homeobox (Arx) was recently demonstrated to be involved in pancreatic alpha cell fate specification while simultaneously repressing the beta and delta cell lineages. To establish whether Arx is not only necessary, but also sufficient to instruct the alpha cell fate in endocrine progenitors, we used a gain-of-function approach to generate mice conditionally misexpressing this factor. Mice with forced Arx expression in the embryonic pancreas or in developing islet cells developed a dramatic hyperglycemia and eventually died. Further analysis demonstrated a drastic loss of beta and delta cells. Concurrently, a remarkable increase in the number of cells displaying alpha cell or, strikingly, pancreatic polypeptide (PP) cell features was observed. Notably, the ectopic expression of Arx induced in embryonic or adult beta cells led to a loss of the beta cell phenotype and a concomitant increase in a number of cells with alpha or PP cell characteristics. Combining quantitative real-time PCR and lineage-tracing experiments, we demonstrate that, in adult mice, the misexpression of Arx, rather than its overexpression, promotes a conversion of beta cells into glucagon- or PP-producing cells in vivo. These results provide important insights into the complex mechanisms underlying proper pancreatic endocrine cell allocation and cell identity acquisition.


Asunto(s)
Proteínas de Homeodominio/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/embriología , Polipéptido Pancreático/metabolismo , Factores de Transcripción/genética , Animales , Regulación de la Expresión Génica , Glucagón/metabolismo , Islotes Pancreáticos/crecimiento & desarrollo , Ratones , Fenotipo , Reacción en Cadena de la Polimerasa
20.
Genome Biol ; 21(1): 106, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375897

RESUMEN

BACKGROUND: Single-cell RNA-seq (scRNA-seq) is emerging as a powerful tool to dissect cell-specific effects of drug treatment in complex tissues. This application requires high levels of precision, robustness, and quantitative accuracy-beyond those achievable with existing methods for mainly qualitative single-cell analysis. Here, we establish the use of standardized reference cells as spike-in controls for accurate and robust dissection of single-cell drug responses. RESULTS: We find that contamination by cell-free RNA can constitute up to 20% of reads in human primary tissue samples, and we show that the ensuing biases can be removed effectively using a novel bioinformatics algorithm. Applying our method to both human and mouse pancreatic islets treated ex vivo, we obtain an accurate and quantitative assessment of cell-specific drug effects on the transcriptome. We observe that FOXO inhibition induces dedifferentiation of both alpha and beta cells, while artemether treatment upregulates insulin and other beta cell marker genes in a subset of alpha cells. In beta cells, dedifferentiation and insulin repression upon artemether treatment occurs predominantly in mouse but not in human samples. CONCLUSIONS: This new method for quantitative, error-correcting, scRNA-seq data normalization using spike-in reference cells helps clarify complex cell-specific effects of pharmacological perturbations with single-cell resolution and high quantitative accuracy.


Asunto(s)
Islotes Pancreáticos/efectos de los fármacos , RNA-Seq/normas , Análisis de la Célula Individual/normas , Animales , Arteméter/farmacología , Desdiferenciación Celular/efectos de los fármacos , Factores de Transcripción Forkhead/antagonistas & inhibidores , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Aprendizaje Automático , Ratones , Estándares de Referencia , Especificidad de la Especie , Transcriptoma/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA