Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 980
Filtrar
1.
Rev. esp. investig. quir ; 25(3): 97-102, 2022. ilus, tab
Artigo em Espanhol | IBECS | ID: ibc-211158

RESUMO

El páncreas es un órgano de origen endodérmico, que se desarrolla de dos esbozos intestinales separados, uno dorsal y otro ventral,a partir de la cuarta o quinta semana de gestación. El páncreas se origina en el intestino anterior en la parte correspondiente a lafutura segunda porción duodenal. Allí se originan dos brotes: uno posterior o dorsal que aparece a principios de la cuarta semanay crece rápidamente en el mesenterio dorsal y el otro brote es anterior o ventral. Este último da origen a dos brotes, uno origina unaparte del páncreas y otro a la vía biliar e hígado. Existen diferentes variantes anatómicas, derivadas de este desarrollo embrionario;su conocimiento es de vital importancia en pacientes con persistencia de dolor abdominal y episodios de pancreatitis recurrente.El objetivo de este artículo es una revisión de las variantes anatómicas del conducto pancreático que pueden manifestarse como pancreatitis idiopática recurrente. (AU)


The pancreas is an organ of endodermal origin, which develops from two separate intestinal sketches, one dorsal and one ventral,from the fourth or fifth week of gestation. The pancreas originates in the anterior intestine in the part corresponding to the futuresecond duodenal portion. There two shoots originate: one posterior or dorsal that appears at the beginning of the fourth week andgrows rapidly in the dorsal mesentery and the other outbreak is anterior or ventral. The latter gives rise to two outbreaks, oneoriginates a part of the pancreas and another to the bile duct and liver. There are different anatomical variants, derived from thisembryonic development; their knowledge is of vital importance in patients with persistent abdominal pain and episodes of recurrentpancreatitis. The objective of this article is a review of the anatomical variants of the pancreatic duct that can manifest as recurrent idiopathic pancreatitis. (AU)


Assuntos
Humanos , Pâncreas/anormalidades , Pâncreas/anatomia & histologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/ultraestrutura , Ductos Pancreáticos/anormalidades , Ductos Pancreáticos/anatomia & histologia , Ductos Pancreáticos/crescimento & desenvolvimento , Ductos Pancreáticos/ultraestrutura
2.
Cell Mol Life Sci ; 78(23): 7107-7132, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34613423

RESUMO

Over the past few years, extensive efforts have been made to generate in-vitro pancreatic micro-tissue, for disease modeling or cell replacement approaches in pancreatic related diseases such as diabetes mellitus. To obtain these goals, a closer look at the diverse cells participating in pancreatic development is necessary. Five major non-epithelial pancreatic (pN-Epi) cell populations namely, pancreatic endothelium, mesothelium, neural crests, pericytes, and stellate cells exist in pancreas throughout its development, and they are hypothesized to be endogenous inducers of the development. In this review, we discuss different pN-Epi cells migrating to and existing within the pancreas and their diverse effects on pancreatic epithelium during organ development mediated via associated signaling pathways, soluble factors or mechanical cell-cell interactions. In-vivo and in-vitro experiments, with a focus on N-Epi cells' impact on pancreas endocrine development, have also been considered. Pluripotent stem cell technology and multicellular three-dimensional organoids as new approaches to generate pancreatic micro-tissues have also been discussed. Main challenges for reaching a detailed understanding of the role of pN-Epi cells in pancreas development in utilizing for in-vitro recapitulation have been summarized. Finally, various novel and innovative large-scale bioengineering approaches which may help to recapitulate cell-cell interactions and are crucial for generation of large-scale in-vitro multicellular pancreatic micro-tissues, are discussed.


Assuntos
Comunicação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Diabetes Mellitus/terapia , Pâncreas/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Diferenciação Celular/fisiologia , Células Endoteliais/metabolismo , Endotélio/citologia , Endotélio/metabolismo , Humanos , Organogênese/fisiologia , Organoides/citologia , Pâncreas/citologia , Pancreatopatias/terapia , Células-Tronco Pluripotentes/citologia
3.
Int. j. morphol ; 39(5): 1459-1466, oct. 2021. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1385497

RESUMO

SUMMARY: This study aimed to investigate the effect of exogenous ghrelin on pancreatic growth and development in African ostrich chicks. Sixteen 40-day-old African ostrich chicks (male or female) were randomly divided into four groups and injected intravenously metatarsal vein with saline (control) or ghrelin (10, 50, and 100 μg/kg) for 6 days. Body and pancreas weight were determined, structural characteristics were observed using HE staining, somatostatin-immunopositive cells were detected using immunohistochemistry. The results were as follows: 1. The 50 and 100 μg/kg groups showed lower relative pancreas weight than the control group (P 0.05. Moreover, compared with the control, the islet cells in treatment groups were loosely arranged and showed reduced cytoplasm. In the exocrine pancreas, the volume of acinar cells in the 10, 50, and 100 μg/kg groups all decreased to varying degrees. 3. Somatostatin immunopositive cells were mainly located around the periphery of the islets and sporadically distributed in the center. The density of the somatostatin immunopositive cells in the 10, 50, and 100 μg/kg groups was higher than that in the control (P < 0.05). These findings suggest that exogenous ghrelin increases the area and number of islets and number of somatostatin immunopositive cells but reduces relative pancreas weight and effects the morphological and structural development of the pancreas, which may inhibit the pancreatic growth and development in African ostrich chicks.


RESUMEN: Este estudio tuvo como objetivo investigar el efecto de la grelina exógena sobre el crecimiento y desarrollo del páncreas en polluelos de avestruz africana. Dieciséis pollos de avestruz africana de 40 días (machos o hembras) se dividieron al azar en cuatro grupos y se inyectaron por vía intravenosa con solución salina (control) o grelina (10, 50 y 100 μg / kg) durante 6 días. determinadas, se observaron las características estructurales mediante tinción Hematoxilina-Eosina, se detectaron células inmunopositivas a somatostatina mediante inmunohistoquímica. Los resultados fueron los siguientes: ¨Los grupos de 50 y 100 μg / kg mostraron un menor peso relativo del páncreas que el grupo de control (P <0,05). El área de islotes por unidad de área del páncreas fue mayor en los grupos de 10, 50 y 100 μg / kg grupos que en el grupo de control (P <0,05). El número de islotes por unidad de área del páncreas fue menor en el grupo de 10 μg / kg que en el control (P <0,05). Además, en comparación con el control, las células de los islotes en los grupos de tratamiento estaban dispuestas de forma holgada y mostraban un citoplasma reducido. En el páncreas exocrino, el volumen de células acinares en los grupos de 10, 50 y 100 μg / kg disminuyó en diversos grados. Las células inmunopositivas de somatostatina se ubicaron principalmente alrededor de la periferia de los islotes y se distribuyeron esporádicamente en el centro. La densidad de las células inmunopositivas a la somatostatina en los grupos de 10, 50 y 100 μg / kg fue mayor que la del control (P <0,05). Estos hallazgos sugieren que la grelina exógena aumenta el área y el número de islotes y el número de células inmunopositivas a la somatostatina, pero reduce el peso relativo del páncreas, lo que puede inhibir el crecimiento y desarrollo pancreático en los polluelos de avestruz africana.


Assuntos
Animais , Pâncreas/efeitos dos fármacos , Struthioniformes , Grelina/administração & dosagem , Pâncreas/crescimento & desenvolvimento , Somatostatina/efeitos dos fármacos , Imuno-Histoquímica , Grelina/farmacologia , Injeções Intravenosas
4.
Exp Mol Pathol ; 122: 104678, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34450114

RESUMO

Stem cell factor (SCF) is an essential cytokine during development and is necessary for gametogenesis, hematopoiesis, mast cell development, stem cell function, and melanogenesis. Here, we measure SCF concentration and distribution in adult humans and mice using gene expression analysis, tissue staining, and organ protein lysates. We demonstrate continued SCF expression in many cell types and tissues into adulthood. Tissues with high expression in adult humans included stomach, spleen, kidney, lung, and pancreas. In mice, we found high SCF expression in the esophagus, ovary, uterus, kidney, and small intestine. Future studies may correlate our findings of increased, organ-specific SCF concentrations within adult tissues with increased risk of SCF/CD117-related disease.


Assuntos
Diferenciação Celular/genética , Proteínas Proto-Oncogênicas c-kit/genética , Fator de Células-Tronco/genética , Distribuição Tecidual/genética , Adulto , Animais , Gametogênese/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematopoese/genética , Humanos , Rim/crescimento & desenvolvimento , Rim/metabolismo , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Mastócitos/metabolismo , Camundongos , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Baço/crescimento & desenvolvimento , Baço/metabolismo , Células-Tronco/metabolismo , Estômago/crescimento & desenvolvimento , Estômago/metabolismo
5.
Development ; 148(16)2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34345920

RESUMO

The melastatin subfamily of the transient receptor potential channels (TRPM) are regulators of pancreatic ß-cell function. TRPM7 is the most abundant islet TRPM channel; however, the role of TRPM7 in ß-cell function has not been determined. Here, we used various spatiotemporal transgenic mouse models to investigate how TRPM7 knockout influences pancreatic endocrine development, proliferation and function. Ablation of TRPM7 within pancreatic progenitors reduced pancreatic size, and α-cell and ß-cell mass. This resulted in modestly impaired glucose tolerance. However, TRPM7 ablation following endocrine specification or in adult mice did not impact endocrine expansion or glucose tolerance. As TRPM7 regulates cell proliferation, we assessed how TRPM7 influences ß-cell hyperplasia under insulin-resistant conditions. ß-Cell proliferation induced by high-fat diet was significantly decreased in TRPM7-deficient ß-cells. The endocrine roles of TRPM7 may be influenced by cation flux through the channel, and indeed we found that TRPM7 ablation altered ß-cell Mg2+ and reduced the magnitude of elevation in ß-cell Mg2+ during proliferation. Together, these findings revealed that TRPM7 controls pancreatic development and ß-cell proliferation, which is likely due to regulation of Mg2+ homeostasis.


Assuntos
Proliferação de Células/genética , Dieta Hiperlipídica , Secreção de Insulina/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Células Cultivadas , Técnicas de Inativação de Genes , Intolerância à Glucose/genética , Homeostase/genética , Magnésio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Cátion TRPM/genética
6.
In Vitro Cell Dev Biol Anim ; 57(6): 587-597, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34212340

RESUMO

Conventional methods for obtaining pancreatic ß cells are based on simulating the embryonic development phase of endocrine cells via hierarchical differentiation of pluripotent stem cells (PSCs). Accordingly, we attempted to modify the protocols for obtaining insulin-secreting cells (ISCs) by sequential differentiation of a human embryonic stem cell (hESC), using the HS181 cell line. Furthermore, we hypothesize that actual pancreatic endocrine cells may arise from trans-differentiation of mature ductal cells after the embryonic developmental stage and throughout the rest of life. According to the hypothesis, ductal cells are trans-differentiated into endocrine and exocrine cells, undergoing a partial epithelial to mesenchymal transition (EMT). To address this issue, we developed two new protocols based on hESC differentiation to obtain ductal cells and then induce EMT in cells to obtain hormone-secreting islet-like cells (HSCs). The ductal (pre-EMT exocrine) cells were then induced to undergo partial EMT by treating with Wnt3a and activin A, in hypoxia. The cell derived from the latter method significantly expressed the main endocrine cell-specific markers and also ß cells, in particular. These experiments not only support our hypothetical model but also offer a promising approach to develop new methods to compensate ß cell depletion in patients with type 1 diabetes mellitus (T1DM). Although this protocol of generating islet-like cells from ductal cells has a potential to treat T1DM, this strategy may be exploited to optimize the function of these cells in an animal model and future clinical applications.


Assuntos
Transdiferenciação Celular/genética , Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Pluripotentes/citologia , Proteína Wnt3A/genética , Técnicas de Cultura de Células , Diferenciação Celular/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células Endócrinas/citologia , Transição Epitelial-Mesenquimal/genética , Células-Tronco Embrionárias Humanas/transplante , Humanos , Insulina/genética , Insulina/metabolismo , Secreção de Insulina/genética , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/transplante , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Células-Tronco Pluripotentes/transplante
7.
Nat Commun ; 12(1): 4458, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294685

RESUMO

The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by ß-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice, Ppy gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon ß-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.


Assuntos
Insulina/biossíntese , Células Secretoras de Polipeptídeo Pancreático/metabolismo , Polipeptídeo Pancreático/metabolismo , Precursores de Proteínas/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Linhagem da Célula/genética , Feminino , Técnicas de Introdução de Genes , Humanos , Células Secretoras de Insulina/classificação , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Polipeptídeo Pancreático/deficiência , Polipeptídeo Pancreático/genética , Células Secretoras de Polipeptídeo Pancreático/classificação , Células Secretoras de Polipeptídeo Pancreático/citologia , Gravidez , RNA-Seq
8.
Front Endocrinol (Lausanne) ; 12: 692596, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34295307

RESUMO

The occurrence of diabetes mellitus is characterized by pancreatic ß cell loss and chronic hyperglycemia. While Type 1 and Type 2 diabetes are the most common types, rarer forms involve mutations affecting a single gene. This characteristic has made monogenic diabetes an interesting disease group to model in vitro using human pluripotent stem cells (hPSCs). By altering the genotype of the original hPSCs or by deriving human induced pluripotent stem cells (hiPSCs) from patients with monogenic diabetes, changes in the outcome of the in vitro differentiation protocol can be analyzed in detail to infer the regulatory mechanisms affected by the disease-associated genes. This approach has been so far applied to a diversity of genes/diseases and uncovered new mechanisms. The focus of the present review is to discuss the latest findings obtained by modeling monogenic diabetes using hPSC-derived pancreatic cells generated in vitro. We will specifically focus on the interpretation of these studies, the advantages and limitations of the models used, and the future perspectives for improvement.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina/citologia , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Humanos , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento
9.
J Endocrinol ; 250(3): R23-R35, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34196608

RESUMO

Since the discovery of insulin 100 years ago, our knowledge and understanding of diabetes have grown exponentially. Specifically, with regards to the genetics underlying diabetes risk, our discoveries have paralleled developments in our understanding of the human genome and our ability to study genomics at scale; these advancements in genetics have both accompanied and led to those in diabetes treatment. This review will explore the timeline and history of gene discovery and how this has coincided with progress in the fields of genomics. Examples of genetic causes of monogenic diabetes are presented and the continuing expansion of allelic series in these genes and the challenges these now cause for diagnostic interpretation along with opportunities for patient stratification are discussed.


Assuntos
Diabetes Mellitus/genética , Células Secretoras de Insulina/fisiologia , Insulina/história , Animais , Diferenciação Celular/genética , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/história , Predisposição Genética para Doença , Genômica/história , História do Século XX , História do Século XXI , Humanos , Insulina/genética , Insulina/uso terapêutico , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo
10.
Curr Opin Genet Dev ; 70: 32-39, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34062490

RESUMO

Diabetes is a group of metabolic disorders, which results from insufficient functional pancreatic ß-cell mass either due to the autoimmune destruction of insulin producing ß-cells, or their death or de-differentiation as compensation for insulin resistance. The ability to reprogram cell types within close developmental proximity to ß-cells offers a strategy to replenish ß-cell mass and a future possible treatment of diabetes. Here, we review recent advances in the fields of pancreas development and lineage reprogramming. We also probe the possibility of using reprogrammed cells as an approach by which to further understand developmental mechanisms, in particular roadblocks to changing cell identity. Finally, we highlight fundamental challenges that need to be overcome to advance lineage reprogramming for generating pancreatic cells.


Assuntos
Reprogramação Celular/fisiologia , Pâncreas/citologia , Animais , Linhagem da Célula , Plasticidade Celular , Técnicas de Reprogramação Celular/métodos , Regulação da Expressão Gênica , Humanos , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Sci Rep ; 11(1): 12614, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131249

RESUMO

In the adult pancreas, the presence of progenitor or stem cells and their potential involvement in homeostasis and regeneration remains unclear. Here, we identify that SET domain-containing protein 4 (SETD4), a histone lysine methyltransferase, is expressed in a small cell population in the adult mouse pancreas. Genetic lineage tracing shows that during pancreatic development, descendants of SETD4+ cells make up over 70% of pancreatic cells and then contribute to each pancreatic lineage during pancreatic homeostasis. SETD4+ cells generate newborn acinar cells in response to cerulein-induced pancreatitis in acinar compartments. Ablation of SETD4+ cells compromises regeneration of acinar cells, in contrast to controls. Our findings provide a new cellular narrative for pancreatic development, homeostasis and response to injury via a small SETD4+ cell population. Potential applications may act to preserve pancreatic function in case of pancreatic disease and/or damage.


Assuntos
Metiltransferases/genética , Pâncreas/metabolismo , Pancreatite/genética , Regeneração/genética , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Linhagem da Célula/genética , Ceruletídeo/toxicidade , Modelos Animais de Doenças , Homeostase/efeitos dos fármacos , Homeostase/genética , Humanos , Camundongos , Pâncreas/crescimento & desenvolvimento , Pâncreas/lesões , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/patologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
12.
Cell Rep ; 35(2): 108981, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852861

RESUMO

Despite the central role of chromosomal context in gene transcription, human noncoding DNA variants are generally studied outside of their genomic location. This limits our understanding of disease-causing regulatory variants. INS promoter mutations cause recessive neonatal diabetes. We show that all INS promoter point mutations in 60 patients disrupt a CC dinucleotide, whereas none affect other elements important for episomal promoter function. To model CC mutations, we humanized an ∼3.1-kb region of the mouse Ins2 gene. This recapitulated developmental chromatin states and cell-specific transcription. A CC mutant allele, however, abrogated active chromatin formation during pancreas development. A search for transcription factors acting through this element revealed that another neonatal diabetes gene product, GLIS3, has a pioneer-like ability to derepress INS chromatin, which is hampered by the CC mutation. Our in vivo analysis, therefore, connects two human genetic defects in an essential mechanism for developmental activation of the INS gene.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Diabetes Mellitus/genética , Insulina/genética , Pâncreas/metabolismo , Mutação Puntual , Proteínas Repressoras/genética , Transativadores/genética , Alelos , Animais , Cromatina/química , Cromatina/patologia , Proteínas de Ligação a DNA/deficiência , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Recém-Nascido , Doenças do Recém-Nascido , Insulina/deficiência , Camundongos , Camundongos Transgênicos , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Regiões Promotoras Genéticas , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Proteínas Repressoras/deficiência , Transativadores/deficiência , Transcrição Gênica
13.
Gene ; 785: 145607, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33775847

RESUMO

Although lifelong renewal and additional compensatory growth in response to demand are undeniable facts, so far, no specific stem cells have been found for pancreatic cells. According to the consensus model, the development of pancreas results from the hierarchical differentiation of pluripotent stem cells towards the appearance of the first endocrine and exocrine cells at approximately 7.5 to 8th gestation week (GW) of human embryo. However, the primitive endocrine cells arising from the embryonic phase of development do not appear to be mature or fully functional. Asymmetric localization of cellular components, such as Numb, partition protein complexes (PAR), planar cell polarity components, and certain mRNAs on the apical and basal sides of epithelial cells, causes cellular polarization. According to our model, the equal distribution of cellular components during symmetric cell division yields similar daughter cells that are associated with duct expansion. In contrast, asymmetric cell division is associated with uneven distribution of cellular components among daughter cells, resulting in different fates. Asymmetric cell division leads to duct branching and the development of acinar and stellate cells by a daughter cell, as well as the development of islet progenitor cells through partial epithelial-to-mesenchymal transition (EMT) and delamination of another daughter cell. Recently, we have developed an efficient method to obtain insulin-secreting cells from the transdifferentiation of hESC-derived ductal cells inducing a partial EMT by treatment with Wnt3A and activin A in a hypoxic environment. Similar models can be offered for other tissues and organs such as mammary glands, lungs, prostate, liver, etc. This model may open a new horizon in the field of regenerative medicine and be useful in explaining the cause of certain abnormalities, such as the occurrence of certain cysts and tumors.


Assuntos
Divisão Celular , Polaridade Celular , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Células-Tronco Pluripotentes/citologia , Animais , Humanos , Modelos Biológicos , Pâncreas/irrigação sanguínea , Pâncreas/embriologia
14.
Development ; 148(6)2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-33653874

RESUMO

To gain a deeper understanding of pancreatic ß-cell development, we used iterative weighted gene correlation network analysis to calculate a gene co-expression network (GCN) from 11 temporally and genetically defined murine cell populations. The GCN, which contained 91 distinct modules, was then used to gain three new biological insights. First, we found that the clustered protocadherin genes are differentially expressed during pancreas development. Pcdhγ genes are preferentially expressed in pancreatic endoderm, Pcdhß genes in nascent islets, and Pcdhα genes in mature ß-cells. Second, after extracting sub-networks of transcriptional regulators for each developmental stage, we identified 81 zinc finger protein (ZFP) genes that are preferentially expressed during endocrine specification and ß-cell maturation. Third, we used the GCN to select three ZFPs for further analysis by CRISPR mutagenesis of mice. Zfp800 null mice exhibited early postnatal lethality, and at E18.5 their pancreata exhibited a reduced number of pancreatic endocrine cells, alterations in exocrine cell morphology, and marked changes in expression of genes involved in protein translation, hormone secretion and developmental pathways in the pancreas. Together, our results suggest that developmentally oriented GCNs have utility for gaining new insights into gene regulation during organogenesis.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Organogênese/genética , Pâncreas/crescimento & desenvolvimento , Animais , Caderinas/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Pâncreas/metabolismo
15.
Development ; 148(6)2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33658226

RESUMO

Groucho-related genes (GRGs) are transcriptional co-repressors that are crucial for many developmental processes. Several essential pancreatic transcription factors are capable of interacting with GRGs; however, the in vivo role of GRG-mediated transcriptional repression in pancreas development is still not well understood. In this study, we used complex mouse genetics and transcriptomic analyses to determine that GRG3 is essential for ß cell development, and in the absence of Grg3 there is compensatory upregulation of Grg4Grg3/4 double mutant mice have severe dysregulation of the pancreas gene program with ectopic expression of canonical liver genes and Foxa1, a master regulator of the liver program. Neurod1, an essential ß cell transcription factor and predicted target of Foxa1, becomes downregulated in Grg3/4 mutants, resulting in reduced ß cell proliferation, hyperglycemia, and early lethality. These findings uncover novel functions of GRG-mediated repression during pancreas development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas Correpressoras/genética , Fator 3-alfa Nuclear de Hepatócito/genética , Pâncreas/crescimento & desenvolvimento , Proteínas Repressoras/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Secretoras de Insulina/metabolismo , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Camundongos , Mutação/genética , Organogênese/genética , Pâncreas/metabolismo
16.
Nat Commun ; 12(1): 1020, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589611

RESUMO

The extracellular matrix (ECM) is unique to each tissue and capable of guiding cell differentiation, migration, morphology, and function. The ECM proteome of different developmental stages has not been systematically studied in the human pancreas. In this study, we apply mass spectrometry-based quantitative proteomics strategies using N,N-dimethyl leucine isobaric tags to delineate proteome-wide and ECM-specific alterations in four age groups: fetal (18-20 weeks gestation), juvenile (5-16 years old), young adults (21-29 years old) and older adults (50-61 years old). We identify 3,523 proteins including 185 ECM proteins and quantify 117 of them. We detect previously unknown proteome and matrisome features during pancreas development and maturation. We also visualize specific ECM proteins of interest using immunofluorescent staining and investigate changes in ECM localization within islet or acinar compartments. This comprehensive proteomics analysis contributes to an improved understanding of the critical roles that ECM plays throughout human pancreas development and maturation.


Assuntos
Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento , Pâncreas/metabolismo , Proteoma/genética , Adolescente , Adulto , Criança , Pré-Escolar , Cromatografia Líquida , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/classificação , Proteínas da Matriz Extracelular/metabolismo , Feminino , Feto , Imunofluorescência , Ontologia Genética , Humanos , Masculino , Pessoa de Meia-Idade , Anotação de Sequência Molecular , Organogênese/genética , Pâncreas/crescimento & desenvolvimento , Proteoma/classificação , Proteoma/metabolismo , Proteômica/métodos , Espectrometria de Massas em Tandem
17.
J Diabetes Investig ; 12(5): 710-727, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33449448

RESUMO

AIMS/INTRODUCTION: ß-Cell dysfunction is a hallmark of type 2 diabetes. In a previous pilot study, we identified an association between genetic variants within the human DACH1 gene and young-onset type 2 diabetes. Here, we characterized the function of dachb, the only dach homologue to be expressed in the pancreas, in developing zebrafish embryos. MATERIALS AND METHODS: We injected one-cell stage embryos with a dachb-morpholino (MO) or with the dachb-MO and dachb messenger ribonucleic acid, and determined the effect on the development of the pancreatic islet. We also carried out quantitative polymerase chain reaction and ribonucleic acid sequencing on the dachb-MO group to determine the effect of dachb knockdown on gene expression. RESULTS: MO-mediated dachb knockdown resulted in impaired islet cell development, with a significant decrease in both the ß-cell and islet cell numbers. This islet developmental defect was rescued when embryos were co-injected with dachb-MO and dachb messenger ribonucleic acid. Knockdown of dachb was associated with a significant downregulation of the ß-cell specific marker gene, insa, and the somatostatin cell marker, sst2, as well as regulators of pancreas development, ptf1a, neuroD, pax6a and nkx6.1, and the cell cycle gene, insm1a. Furthermore, ribonucleic sequencing analysis showed an upregulation of genes enriched in the forkhead box O and mitogen-activated protein kinase signaling pathways in the dachb-MO group, when compared with the control groups. CONCLUSIONS: Together, our results suggest the possible role of dachb in islet development in zebrafish.


Assuntos
Diferenciação Celular/genética , Ilhotas Pancreáticas/embriologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/genética , Animais , Desenvolvimento Embrionário/genética , Expressão Gênica/genética , Morfolinos , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento
18.
Mol Biol Rep ; 48(2): 1615-1623, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33484392

RESUMO

Differentiation processes for generating pancreatic progenitors from pluripotent stem cells inhibit Sonic hedgehog signaling through synthetic antagonists. However, the effect of sonic hedgehog inhibition in differentiating human embryonic stem cells remains unclear. The primary aim of this study was to understand the effect of Sonic hedgehog inhibition on key pancreas-specific transcription factors during differentiation of human embryonic stem cells towards a pancreatic lineage. We differentiated human embryonic stem (ES) cells towards the pancreatic progenitor stage. To analyze the effect of Sonic hedgehog inhibition, we differentiated human ES cells in the presence or absence of pathway antagonist, cyclopamine, using the same concentration (0.25 µM) as reported earlier. Changes in gene expression between the groups were examined by quantitative reverse-transcription PCR and immunoblot analyses. Surprisingly, we found that expression of key transcription factors, PDX1 and SOX9, was not majorly affected by inhibition of Sonic hedgehog signals. Effects of inhibiting Hedgehog signals on pancreas-specific markers in differentiating human embryonic stem cells were analyzed in the study. We identified that the expression of pancreas-specific PDX1 and SOX9 was not affected by the Sonic hedgehog pathway in pancreatic progenitor populations from human ES cells. Thus, the restrictive nature of Hedgehog signaling during the early stages of pancreas formation could be facilitated through a transcriptional network beyond PDX1 and SOX9.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Células-Tronco Embrionárias Humanas/citologia , Fatores de Transcrição SOX9/genética , Transativadores/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/genética , Humanos , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Transdução de Sinais/genética , Fatores de Transcrição/genética
19.
J Cell Physiol ; 236(7): 5253-5264, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33368314

RESUMO

Hypercholesterolemia has been found to be closely linked with a significant increase in both cancer incidence and mortality. However, the exact correlation between serum cholesterol levels and cancer has not been completely deciphered. Here we analyzed the effect of low-density lipoprotein (LDL) cholesterol on prostate and pancreatic cancer cells. We noted that LDL induced a substantial STAT3 activation and JAK1, JAK2, Src activation in diverse prostate and pancreatic tumor cells. Moreover, LDL promoted cancer cell proliferation, migration, and invasion as well as upregulated the expression of diverse oncogenic gene products. However, deletion of LDL-activated STAT3 in LNCaP and PANC-1 cells and reduced LDL-induced cell viability. Simvastatin (SV) treatment also alleviated LDL-induced cell viability and migration ability in both the prostate and pancreatic tumor cells. These results demonstrate that LDL-induced STAT3 activation may exert a profound effect on the proliferation and survival of tumor cells.


Assuntos
Carcinogênese/patologia , LDL-Colesterol/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias da Próstata/patologia , Fator de Transcrição STAT3/metabolismo , Anticolesterolemiantes/farmacologia , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Masculino , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Próstata/citologia , Próstata/crescimento & desenvolvimento , Próstata/patologia , Interferência de RNA , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/fisiologia , Sinvastatina/farmacologia
20.
Cell Mol Life Sci ; 78(1): 287-298, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32189007

RESUMO

Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimulates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk, Jnk3, Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3, CCND1 and CCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we find that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.


Assuntos
Células Secretoras de Insulina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D2/genética , Ciclina D2/metabolismo , Feminino , Glucose/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Obesidade/metabolismo , Obesidade/patologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...