Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37373094

RESUMEN

Adult pancreatic acinar cells show high plasticity allowing them to change in their differentiation commitment. Pancreatic acinar-to-ductal metaplasia (ADM) is a cellular process in which the differentiated pancreatic acinar cells transform into duct-like cells. This process can occur as a result of cellular injury or inflammation in the pancreas. While ADM is a reversible process allowing pancreatic acinar regeneration, persistent inflammation or injury can lead to the development of pancreatic intraepithelial neoplasia (PanIN), which is a common precancerous lesion that precedes pancreatic ductal adenocarcinoma (PDAC). Several factors can contribute to the development of ADM and PanIN, including environmental factors such as obesity, chronic inflammation and genetic mutations. ADM is driven by extrinsic and intrinsic signaling. Here, we review the current knowledge on the cellular and molecular biology of ADM. Understanding the cellular and molecular mechanisms underlying ADM is critical for the development of new therapeutic strategies for pancreatitis and PDAC. Identifying the intermediate states and key molecules that regulate ADM initiation, maintenance and progression may help the development of novel preventive strategies for PDAC.


Asunto(s)
Carcinoma in Situ , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adulto , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Páncreas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Células Acinares/patología , Carcinoma in Situ/genética , Metaplasia/patología , Inflamación/patología , Neoplasias Pancreáticas
2.
J Pathol ; 254(1): 31-45, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33527355

RESUMEN

Maturity-onset diabetes of the young type 5 (MODY5) is due to heterozygous mutations or deletion of HNF1B. No mouse models are currently available to recapitulate the human MODY5 disease. Here, we investigate the pancreatic phenotype of a unique MODY5 mouse model generated by heterozygous insertion of a human HNF1B splicing mutation at the intron-2 splice donor site in the mouse genome. This Hnf1bsp2/+ model generated with targeted mutation of Hnf1b mimicking the c.544+1G>T (T) mutation identified in humans, results in alternative transcripts and a 38% decrease of native Hnf1b transcript levels. As a clinical feature of MODY5 patients, the hypomorphic mouse model Hnf1bsp2/+ displays glucose intolerance. Whereas Hnf1bsp2/+ isolated islets showed no altered insulin secretion, we found a 65% decrease in pancreatic insulin content associated with a 30% decrease in total large islet volume and a 20% decrease in total ß-cell volume. These defects were associated with a 30% decrease in expression of the pro-endocrine gene Neurog3 that we previously identified as a direct target of Hnf1b, showing a developmental etiology. As another clinical feature of MODY5 patients, the Hnf1bsp2/+ pancreases display exocrine dysfunction with hypoplasia. We observed chronic pancreatitis with loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis, with upregulation of signaling pathways and impaired acinar cell regeneration. This was associated with ductal cell deficiency characterized by shortened primary cilia. Importantly, the Hnf1bsp2/+ mouse model reproduces the pancreatic features of the human MODY5/HNF1B disease, providing a unique in vivo tool for molecular studies of the endocrine and exocrine defects and to advance basic and translational research. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/fisiopatología , Esmalte Dental/anomalías , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Factor Nuclear 1-beta del Hepatocito/genética , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/fisiopatología , Páncreas/fisiopatología , Animales , Enfermedades del Sistema Nervioso Central/patología , Esmalte Dental/patología , Esmalte Dental/fisiopatología , Diabetes Mellitus Tipo 2/patología , Humanos , Enfermedades Renales Quísticas/patología , Ratones , Ratones Transgénicos , Mutación , Páncreas/patología , Fenotipo
3.
Cell Mol Gastroenterol Hepatol ; 8(3): 487-511, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31229598

RESUMEN

BACKGROUND & AIMS: The exocrine pancreas consists of acinar cells that produce digestive enzymes transported to the intestine through a branched ductal epithelium. Chronic pancreatitis is characterized by progressive inflammation, fibrosis, and loss of acinar tissue. These changes of the exocrine tissue are risk factors for pancreatic cancer. The cause of chronic pancreatitis cannot be identified in one quarter of patients. Here, we investigated how duct dysfunction could contribute to pancreatitis development. METHODS: The transcription factor Hnf1b, first expressed in pancreatic progenitors, is strictly restricted to ductal cells from late embryogenesis. We previously showed that Hnf1b is crucial for pancreas morphogenesis but its postnatal role still remains unelucidated. To investigate the role of pancreatic ducts in exocrine homeostasis, we inactivated the Hnf1b gene in vivo in mouse ductal cells. RESULTS: We uncovered that postnatal Hnf1b inactivation in pancreatic ducts leads to chronic pancreatitis in adults. Hnf1bΔduct mutants show dilatation of ducts, loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis. We deciphered the early events involved, with down-regulation of cystic disease-associated genes, loss of primary cilia, up-regulation of signaling pathways, especially the Yap pathway, which is involved in acinar-to-ductal metaplasia. Remarkably, Hnf1bΔduct mutants developed pancreatic intraepithelial neoplasia and promote pancreatic intraepithelial neoplasia progression in concert with KRAS. We further showed that adult Hnf1b inactivation in pancreatic ducts is associated with impaired regeneration after injury, with persistent metaplasia and initiation of neoplasia. CONCLUSIONS: Loss of Hnf1b in ductal cells leads to chronic pancreatitis and neoplasia. This study shows that Hnf1b deficiency may contribute to diseases of the exocrine pancreas and gains further insight into the etiology of pancreatitis and tumorigenesis.


Asunto(s)
Carcinoma in Situ/genética , Eliminación de Gen , Factor Nuclear 1-beta del Hepatocito/genética , Conductos Pancreáticos/crecimiento & desarrollo , Neoplasias Pancreáticas/genética , Pancreatitis/genética , Animales , Animales Recién Nacidos , Carcinoma in Situ/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Factor Nuclear 1-beta del Hepatocito/metabolismo , Homeostasis , Humanos , Ratones , Páncreas Exocrino/metabolismo , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Pancreatitis/complicaciones , Pancreatitis/metabolismo , Transducción de Señal
4.
Br J Pharmacol ; 174(21): 3865-3880, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28832971

RESUMEN

BACKGROUND AND PURPOSE: Pancreatitis is a common inflammation of the pancreas with rising incidence in many countries. Despite improvements in diagnostic techniques, the disease is associated with high risk of severe morbidity and mortality and there is an urgent need for new therapeutic interventions. In this study, we evaluated whether histone deacetylases (HDACs), key epigenetic regulators of gene transcription, are involved in the development of the disease. EXPERIMENTAL APPROACH: We analysed HDAC regulation during cerulein-induced acute, chronic and autoimmune pancreatitis using different transgenic mouse models. The functional relevance of class I HDACs was tested with the selective inhibitor MS-275 in vivo upon pancreatitis induction and in vitro in activated macrophages and primary acinar cell explants. KEY RESULTS: HDAC expression and activity were up-regulated in a time-dependent manner following induction of pancreatitis, with the highest abundance observed for class I HDACs. Class I HDAC inhibition did not prevent the initial acinar cell damage. However, it effectively reduced the infiltration of inflammatory cells, including macrophages and T cells, in both acute and chronic phases of the disease, and directly disrupted macrophage activation. In addition, MS-275 treatment reduced DNA damage in acinar cells and limited acinar de-differentiation into acinar-to-ductal metaplasia in a cell-autonomous manner by impeding the EGF receptor signalling axis. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that class I HDACs are critically involved in the development of acute and chronic forms of pancreatitis and suggest that blockade of class I HDAC isoforms is a promising target to improve the outcome of the disease.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , Pancreatitis/tratamiento farmacológico , Células Acinares/metabolismo , Enfermedad Aguda , Animales , Enfermedades Autoinmunes/fisiopatología , Benzamidas/farmacología , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Histona Desacetilasas/metabolismo , Leucocitos/metabolismo , Masculino , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pancreatitis/fisiopatología , Pancreatitis Crónica/tratamiento farmacológico , Pancreatitis Crónica/fisiopatología , Piridinas/farmacología , Factores de Tiempo
5.
Best Pract Res Clin Endocrinol Metab ; 29(6): 883-98, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26696517

RESUMEN

Pancreas development is controlled by a complex interaction of signaling pathways and transcription factor networks that determine pancreatic specification and differentiation of exocrine and endocrine cells. Epigenetics adds a new layer of gene regulation. DNA methylation, histone modifications and non-coding RNAs recently appeared as important epigenetic factors regulating pancreas development. In this review, we report recent findings obtained by analyses in model organisms as well as genome-wide approaches that demonstrate the role of these epigenetic regulators in the control of exocrine and endocrine cell differentiation, identity, function, proliferation and regeneration. We also highlight how altered epigenetic processes contribute to pancreatic disorders: diabetes and pancreatic cancer. Uncovering these epigenetic events can help to better understand these diseases, provide novel therapeutical targets for their treatment, and improve cell-based therapies for diabetes.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/genética , Epigénesis Genética , Páncreas/citología , Neoplasias Pancreáticas/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo
6.
Development ; 142(5): 871-82, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25715395

RESUMEN

Heterozygous mutations in the human HNF1B gene are associated with maturity-onset diabetes of the young type 5 (MODY5) and pancreas hypoplasia. In mouse, Hnf1b heterozygous mutants do not exhibit any phenotype, whereas the homozygous deletion in the entire epiblast leads to pancreas agenesis associated with abnormal gut regionalization. Here, we examine the specific role of Hnf1b during pancreas development, using constitutive and inducible conditional inactivation approaches at key developmental stages. Hnf1b early deletion leads to a reduced pool of pancreatic multipotent progenitor cells (MPCs) due to decreased proliferation and increased apoptosis. Lack of Hnf1b either during the first or the secondary transitions is associated with cystic ducts. Ductal cells exhibit aberrant polarity and decreased expression of several cystic disease genes, some of which we identified as novel Hnf1b targets. Notably, we show that Glis3, a transcription factor involved in duct morphogenesis and endocrine cell development, is downstream Hnf1b. In addition, a loss and abnormal differentiation of acinar cells are observed. Strikingly, inactivation of Hnf1b at different time points results in the absence of Ngn3(+) endocrine precursors throughout embryogenesis. We further show that Hnf1b occupies novel Ngn3 putative regulatory sequences in vivo. Thus, Hnf1b plays a crucial role in the regulatory networks that control pancreatic MPC expansion, acinar cell identity, duct morphogenesis and generation of endocrine precursors. Our results uncover an unappreciated requirement of Hnf1b in endocrine cell specification and suggest a mechanistic explanation of diabetes onset in individuals with MODY5.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Páncreas/citología , Páncreas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Conducto Cístico/citología , Conducto Cístico/metabolismo , Proteínas de Unión al ADN , Femenino , Factor Nuclear 1-beta del Hepatocito/genética , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Proteínas del Tejido Nervioso/genética , Embarazo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
7.
Cell Cycle ; 8(4): 536-44, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19197155

RESUMEN

Genes in the mammalian genome contain information necessary to build an organism during development. Epigenetic processes add a further degree of complexity. These mechanisms of temporal and spatial control of gene activity during the development of complex organisms modulate gene expression patterns without modifying the DNA sequence. Post-translational modifications of histones such as acetylation bestow the ability to modify genomic signals. Determining whether epigenetic changes are responsible for particular phenotypes is thus crucial to understand organ development. Here we review the role of histone deacetylase enzymes (HDACs) in guiding lineage commitment and driving cell differentiation, as well as their pharmacological manipulation using small-molecule HDAC inhibitors in various differentiation programs. In particular, we focus on the pancreas as we recently discovered that deacetylase inhibition favors generation of endocrine pancreatic cells. We also discuss the potential application of HDAC inhibitors for disease treatment, with particular emphasis on diabetes therapy.


Asunto(s)
Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Animales , Encefalopatías/enzimología , Encefalopatías/fisiopatología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula , Células Endocrinas/citología , Células Endocrinas/fisiología , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Histona Desacetilasas/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Páncreas/citología , Páncreas/fisiología
8.
Mol Cell Biol ; 28(20): 6373-83, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18710955

RESUMEN

During pancreas development, transcription factors play critical roles in exocrine and endocrine differentiation. Transcriptional regulation in eukaryotes occurs within chromatin and is influenced by posttranslational histone modifications (e.g., acetylation) involving histone deacetylases (HDACs). Here, we show that HDAC expression and activity are developmentally regulated in the embryonic rat pancreas. We discovered that pancreatic treatment with different HDAC inhibitors (HDACi) modified the timing and determination of pancreatic cell fate. HDACi modified the exocrine lineage via abolition and enhancement of acinar and ductal differentiation, respectively. Importantly, HDACi treatment promoted the NGN3 proendocrine lineage, leading to an increased pool of endocrine progenitors and modified endocrine subtype lineage choices. Interestingly, treatments with trichostatin A and sodium butyrate, two inhibitors of both class I and class II HDACs, enhanced the pool of beta cells. These results highlight the roles of HDACs at key points in exocrine and endocrine differentiation. They show the powerful use of HDACi to switch pancreatic cell determination and amplify specific cellular subtypes, with potential applications in cell replacement therapies in diabetes.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Islotes Pancreáticos/citología , Páncreas/efectos de los fármacos , Páncreas/enzimología , Células Madre/citología , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Ácidos Hidroxámicos/farmacología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/enzimología , Proteínas del Tejido Nervioso/metabolismo , Páncreas/citología , Páncreas/embriología , Páncreas Exocrino/citología , Páncreas Exocrino/efectos de los fármacos , Páncreas Exocrino/enzimología , Conductos Pancreáticos/citología , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/enzimología , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/efectos de los fármacos , Células Secretoras de Polipéptido Pancreático/enzimología , Ratas , Ratas Wistar , Células Madre/efectos de los fármacos , Ácido Valproico/farmacología
9.
Hum Mol Genet ; 15(15): 2363-75, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16801329

RESUMEN

Heterozygous mutations in the HNF1beta/vHNF1/TCF2 gene cause maturity-onset diabetes of the young (MODY5), associated with severe renal disease and abnormal genital tract. Here, we characterize two fetuses, a 27-week male and a 31.5-week female, carrying novel mutations in exons 2 and 7 of HNF1beta, respectively. Although these mutations were predicted to have different functional consequences, both fetuses displayed highly similar phenotypes. They presented one of the most severe phenotypes described in HNF1beta carriers: bilateral enlarged polycystic kidneys, severe pancreas hypoplasia and abnormal genital tract. Consistent with this, we detected high levels of HNF1beta transcripts in 8-week human embryos in the mesonephros and metanephric kidney and in the epithelium of pancreas. Renal histology and immunohistochemistry analyses of mutant fetuses revealed cysts derived from all nephron segments with multilayered epithelia and dysplastic regions, accompanied by a marked increase in the expression of beta-catenin and E-cadherin. A significant proportion of cysts still expressed the cystic renal disease proteins, polycystin-1, polycystin-2, fibrocystin and uromodulin, implying that cyst formation may result from a deregulation of cell-cell adhesion and/or the Wnt/beta-catenin signaling pathway. Both fetuses exhibited a severe pancreatic hypoplasia with underdeveloped and disorganized acini, together with an absence of ventral pancreatic-derived tissue. beta-catenin and E-cadherin were strongly downregulated in the exocrine and endocrine compartments, and the islets lacked the transporter essential for glucose-sensing GLUT2, indicating a beta-cell maturation defect. This study provides evidence of differential gene-dosage requirements for HNF1beta in normal human kidney and pancreas differentiation and increases our understanding of the etiology of MODY5 disorder.


Asunto(s)
Enfermedades Fetales/genética , Mutación del Sistema de Lectura , Factor Nuclear 1-beta del Hepatocito/genética , Riñón Displástico Multiquístico/genética , Páncreas/anomalías , Feto Abortado , Adulto , Diabetes Mellitus Tipo 2/embriología , Diabetes Mellitus Tipo 2/genética , Epitelio/metabolismo , Epitelio/patología , Femenino , Dosificación de Gen/fisiología , Factor Nuclear 1-beta del Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Riñón/embriología , Riñón/metabolismo , Riñón/patología , Masculino , Riñón Displástico Multiquístico/diagnóstico , Páncreas/embriología , Páncreas/ultraestructura , Embarazo
10.
J Biol Chem ; 278(42): 40933-42, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12860991

RESUMEN

The two members of the hepatocyte nuclear factor 1 (HNF1) transcription factor family, HNF1 and variant HNF1 (vHNF1), show a strong homology in their atypical POU-homeodomain and dimerization domain but differ in their transactivation domains. Moreover, two vHNF1 isoforms generated by alternative splicing are present in all tissues expressing this gene. vHnf1-deficient mouse embryos die soon after implantation due to defective visceral endoderm formation, an extraembryonic tissue essential for development and survival of the embryo proper. In contrast, invalidation of Hnf1, which is expressed at later developmental stages than vHnf1, does not lead to embryonic lethality or developmental defects. To examine the specific or potential equivalent functions of vHNF1 isoforms and HNF1 during the process of visceral endoderm differentiation, we stably reexpressed these factors in vHnf1-deficient embryonic stem cells. Analysis of these embryonic stem cells upon differentiation into embryoid bodies shows that vHNF1 isoforms exhibit specific behaviors depending on particular target genes and cooperate in the establishment of a functional visceral endoderm. Furthermore, forced expression of HNF1 in vHnf1-deficient embryonic stem cells fully restores the formation of a mature visceral endoderm with the correct expression profile of early and late markers of this lineage. Thus, in this context, HNF1 functionally replaces both vHNF1 isoforms, suggesting that the different developmental functions of these transcription factors are mainly due to the acquisition of novel expression patterns.


Asunto(s)
Proteínas de Unión al ADN , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares , Factores de Transcripción/fisiología , Empalme Alternativo , Animales , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Linaje de la Célula , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electroporación , Factor Nuclear 1 del Hepatocito , Factor Nuclear 1-alfa del Hepatocito , Factor Nuclear 1-beta del Hepatocito , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Plásmidos/metabolismo , Isoformas de Proteínas , Estructura Terciaria de Proteína , ARN/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional , Transfección , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA