Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Mol Gastroenterol Hepatol ; 18(2): 101347, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670488

RESUMEN

BACKGROUND & AIM: Telocytes, a recently identified type of subepithelial interstitial cell, have garnered attention for their potential roles in tissue homeostasis and repair. However, their contribution to gastric metaplasia remains unexplored. This study elucidates the role of telocytes in the development of metaplasia within the gastric environment. METHODS: To investigate the presence and behavior of telocytes during metaplastic transitions, we used drug-induced acute injury models (using DMP-777 or L635) and a genetically engineered mouse model (Mist1-Kras). Lineage tracing via the Foxl1-CreERT2;R26R-tdTomato mouse model was used to track telocyte migratory dynamics. Immunofluorescence staining was used to identify telocyte markers and evaluate their correlation with metaplasia-related changes. RESULTS: We confirmed the existence of FOXL1+/PDGFRα+ double-positive telocytes in the stomach's isthmus region. As metaplasia developed, we observed a marked increase in the telocyte population. The distribution of telocytes expanded beyond the isthmus to encompass the entire gland and closely reflected the expansion of the proliferative cell zone. Rather than a general response to mucosal damage, the shift in telocyte distribution was associated with the establishment of a metaplastic cell niche at the gland base. Furthermore, lineage-tracing experiments highlighted the active recruitment of telocytes to the emerging metaplastic cell niche, and we observed expression of Wnt5a, Bmp4, and Bmp7 in PDGFRα+ telocytes. CONCLUSIONS: These results suggest that telocytes contribute to the evolution of a gastric metaplasia niche. The dynamic behavior of these stromal cells, their responsiveness to metaplastic changes, and potential association with Wnt5a, Bmp4, and Bmp7 signaling emphasize the significance of telocytes in tissue adaptation and repair.


Asunto(s)
Proteína Morfogenética Ósea 4 , Mucosa Gástrica , Metaplasia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Telocitos , Proteína Wnt-5a , Animales , Metaplasia/patología , Ratones , Telocitos/metabolismo , Telocitos/patología , Proteína Wnt-5a/metabolismo , Mucosa Gástrica/patología , Mucosa Gástrica/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estómago/patología , Proteína Morfogenética Ósea 7/metabolismo , Movimiento Celular , Ratones Transgénicos , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead
2.
Stem Cells ; 41(6): 643-657, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37085274

RESUMEN

During early embryogenesis, the transcription factor SOX17 contributes to hepato-pancreato-biliary system formation and vascular-hematopoietic emergence. To better understand Sox17 function in the developing endoderm and endothelium, we developed a dual-color temporal lineage-tracing strategy in mice combined with single-cell RNA sequencing to analyze 6934 cells from Sox17-expressing lineages at embryonic days 9.0-9.5. Our analyses showed 19 distinct cellular clusters combined from all 3 germ layers. Differential gene expression, trajectory and RNA-velocity analyses of endothelial cells revealed a heterogenous population of uncommitted and specialized endothelial subtypes, including 2 hemogenic populations that arise from different origins. Similarly, analyses of posterior foregut endoderm revealed subsets of hepatic, pancreatic, and biliary progenitors with overlapping developmental potency. Calculated gene-regulatory networks predict gene regulons that are dominated by cell type-specific transcription factors unique to each lineage. Vastly different Sox17 regulons found in endoderm versus endothelial cells support the differential interactions of SOX17 with other regulatory factors thereby enabling lineage-specific regulatory actions.


Asunto(s)
Desarrollo Embrionario , Células Endoteliales , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Factores de Transcripción SOXF , Animales , Ratones , Diferenciación Celular , Linaje de la Célula/genética , Endodermo/metabolismo , Células Endoteliales/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Análisis de Secuencia de ARN , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción/metabolismo , Desarrollo Embrionario/genética
4.
Dev Cell ; 53(4): 390-405.e10, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32359405

RESUMEN

Although cellular stress response is important for maintaining function and survival, overactivation of late-stage stress effectors cause dysfunction and death. We show that the myelin transcription factors (TFs) Myt1 (Nzf2), Myt2 (Myt1l, Nztf1, and Png-1), and Myt3 (St18 and Nzf3) prevent such overactivation in islet ß cells. Thus, we found that co-inactivating the Myt TFs in mouse pancreatic progenitors compromised postnatal ß cell function, proliferation, and survival, preceded by upregulation of late-stage stress-response genes activating transcription factors (e.g., Atf4) and heat-shock proteins (Hsps). Myt1 binds putative enhancers of Atf4 and Hsps, whose overexpression largely recapitulated the Myt-mutant phenotypes. Moreover, Myt(MYT)-TF levels were upregulated in mouse and human ß cells during metabolic stress-induced compensation but downregulated in dysfunctional type 2 diabetic (T2D) human ß cells. Lastly, MYT knockdown caused stress-gene overactivation and death in human EndoC-ßH1 cells. These findings suggest that Myt TFs are essential restrictors of stress-response overactivity.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Diabetes Mellitus/patología , Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/citología , Estrés Fisiológico , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Factor de Transcripción Activador 4/genética , Animales , Proliferación Celular , Proteínas de Unión al ADN/genética , Diabetes Mellitus/metabolismo , Femenino , Proteínas de Choque Térmico/genética , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción/genética
5.
Diabetes ; 69(6): 1219-1231, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32245798

RESUMEN

Swi-independent 3a and 3b (Sin3a and Sin3b) are paralogous transcriptional coregulators that direct cellular differentiation, survival, and function. Here, we report that mouse Sin3a and Sin3b are coproduced in most pancreatic cells during embryogenesis but become much more enriched in endocrine cells in adults, implying continued essential roles in mature endocrine cell function. Mice with loss of Sin3a in endocrine progenitors were normal during early postnatal stages but gradually developed diabetes before weaning. These physiological defects were preceded by the compromised survival, insulin-vesicle packaging, insulin secretion, and nutrient-induced Ca2+ influx of Sin3a-deficient ß-cells. RNA sequencing coupled with candidate chromatin immunoprecipitation assays revealed several genes that could be directly regulated by Sin3a in ß-cells, which modulate Ca2+/ion transport, cell survival, vesicle/membrane trafficking, glucose metabolism, and stress responses. Finally, mice with loss of both Sin3a and Sin3b in multipotent embryonic pancreatic progenitors had significantly reduced islet cell mass at birth, caused by decreased endocrine progenitor production and increased ß-cell death. These findings highlight the stage-specific requirements for the presumed "general" coregulators Sin3a and Sin3b in islet ß-cells, with Sin3a being dispensable for differentiation but required for postnatal function and survival.


Asunto(s)
Calcio/metabolismo , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/fisiología , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Envejecimiento , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Supervivencia Celular , Diabetes Mellitus/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/genética , Complejo Correpresor Histona Desacetilasa y Sin3/genética
7.
Dev Cell ; 50(6): 744-754.e4, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31422917

RESUMEN

Activating mutations in Kras are nearly ubiquitous in human pancreatic cancer and initiate precancerous pancreatic intraepithelial neoplasia (PanINs) when induced in mouse acinar cells. PanINs normally take months to form but are accelerated by deletion of acinar cell differentiation factors such as Ptf1a, suggesting that loss of cell identity is rate limiting for pancreatic tumor initiation. Using a genetic mouse model that allows for independent control of oncogenic Kras and Ptf1a expression, we demonstrate that sustained Ptf1a is sufficient to prevent Kras-driven tumorigenesis, even in the presence of tumor-promoting inflammation. Furthermore, reintroducing Ptf1a into established PanINs reverts them to quiescent acinar cells in vivo. Similarly, Ptf1a re-expression in human pancreatic cancer cells inhibits their growth and colony-forming ability. Our results suggest that reactivation of an endogenous differentiation program can prevent and reverse oncogene-driven transformation in cells harboring tumor-driving mutations, introducing a potential paradigm for solid tumor prevention and treatment.


Asunto(s)
Carcinogénesis/patología , Diferenciación Celular , Neoplasias Pancreáticas/patología , Células Acinares/metabolismo , Células Acinares/patología , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/patología , Ratones , Neoplasias Pancreáticas/genética , Pancreatitis/patología , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
8.
Curr Biol ; 29(14): 2339-2350.e5, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31303487

RESUMEN

The microtubule (MT) network is an essential regulator of insulin secretion from pancreatic ß cells, which is central to blood-sugar homeostasis. We find that when glucose metabolism induces insulin secretion, it also increases formation of Golgi-derived microtubules (GDMTs), notably with the same biphasic kinetics as insulin exocytosis. Furthermore, GDMT nucleation is controlled by a glucose signal-transduction pathway through cAMP and its effector EPAC2. Preventing new GDMT nucleation dramatically affects the pipeline of insulin production, storage, and release. There is an overall reduction of ß-cell insulin content, and remaining insulin becomes retained within the Golgi, likely because of stalling of insulin-granule budding. While not preventing glucose-induced insulin exocytosis, the diminished granule availability substantially blunts the amount secreted. Constant dynamic maintenance of the GDMT network is therefore critical for normal ß-cell physiology. Our study demonstrates that the biogenesis of post-Golgi carriers, particularly large secretory granules, requires ongoing nucleation and replenishment of the GDMT network.


Asunto(s)
AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Secretoras de Insulina/fisiología , Microtúbulos/metabolismo , Biogénesis de Organelos , Vesículas Secretoras/metabolismo , Animales , Glucosa/metabolismo , Aparato de Golgi/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR
9.
Diabetes ; 68(9): 1806-1818, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31201281

RESUMEN

Transcription factors positively and/or negatively impact gene expression by recruiting coregulatory factors, which interact through protein-protein binding. Here we demonstrate that mouse pancreas size and islet ß-cell function are controlled by the ATP-dependent Swi/Snf chromatin remodeling coregulatory complex that physically associates with Pdx1, a diabetes-linked transcription factor essential to pancreatic morphogenesis and adult islet cell function and maintenance. Early embryonic deletion of just the Swi/Snf Brg1 ATPase subunit reduced multipotent pancreatic progenitor cell proliferation and resulted in pancreas hypoplasia. In contrast, removal of both Swi/Snf ATPase subunits, Brg1 and Brm, was necessary to compromise adult islet ß-cell activity, which included whole-animal glucose intolerance, hyperglycemia, and impaired insulin secretion. Notably, lineage-tracing analysis revealed Swi/Snf-deficient ß-cells lost the ability to produce the mRNAs for Ins and other key metabolic genes without effecting the expression of many essential islet-enriched transcription factors. Swi/Snf was necessary for Pdx1 to bind to the Ins gene enhancer, demonstrating the importance of this association in mediating chromatin accessibility. These results illustrate how fundamental the Pdx1:Swi/Snf coregulator complex is in the pancreas, and we discuss how disrupting their association could influence type 1 and type 2 diabetes susceptibility.


Asunto(s)
Proliferación Celular/fisiología , Ensamble y Desensamble de Cromatina/fisiología , ADN Helicasas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Nucleares/metabolismo , Páncreas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , ADN Helicasas/genética , Regulación de la Expresión Génica , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Proteínas de Homeodominio/genética , Insulina/sangre , Células Secretoras de Insulina/citología , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Páncreas/citología , Transactivadores/genética , Factores de Transcripción/genética
10.
Cell Mol Gastroenterol Hepatol ; 8(2): 173-192, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31125624

RESUMEN

BACKGROUND & AIMS: Pancreatitis is a major cause of morbidity and mortality and is a risk factor for pancreatic tumorigenesis. Upon tissue damage, an inflammatory response, made up largely of macrophages, provides multiple growth factors that promote repair. Here, we examine the molecular pathways initiated by macrophages to promote pancreas recovery from pancreatitis. METHODS: To induce organ damage, mice were subjected to cerulein-induced experimental pancreatitis and analyzed at various times of recovery. CD11b-DTR mice were used to deplete myeloid cells. Hbegff/f;LysM-Cre mice were used to ablate myeloid cell-derived heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF). To ablate EGFR specifically during recovery, pancreatitis was induced in Egfrf/f;Ptf1aFlpO/+;FSF-Rosa26CAG-CreERT2 mice followed by tamoxifen treatment. RESULTS: Macrophages infiltrating the pancreas in experimental pancreatitis make high levels of HB-EGF. Both depletion of myeloid cells and ablation of myeloid cell HB-EGF delayed recovery from experimental pancreatitis, resulting from a decrease in cell proliferation and an increase in apoptosis. Mechanistically, ablation of myeloid cell HB-EGF impaired epithelial cell DNA repair, ultimately leading to cell death. Soluble HB-EGF induced EGFR nuclear translocation and methylation of histone H4, facilitating resolution of DNA damage in pancreatic acinar cells in vitro. Consistent with its role as the primary receptor of HB-EGF, in vivo ablation of EGFR from pancreatic epithelium during recovery from pancreatitis resulted in accumulation of DNA damage. CONCLUSIONS: By using novel conditional knockout mouse models, we determined that HB-EGF derived exclusively from myeloid cells induces epithelial cell proliferation and EGFR-dependent DNA repair, facilitating pancreas healing after injury.


Asunto(s)
Reparación del ADN , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Células Mieloides/metabolismo , Páncreas/fisiología , Pancreatitis/fisiopatología , Regeneración , Animales , ADN/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/farmacología , Ratones , Ratones Noqueados
11.
Mol Metab ; 24: 80-97, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30930126

RESUMEN

OBJECTIVE: Hundreds of missense mutations in the coding region of PDX1 exist; however, if these mutations predispose to diabetes mellitus is unknown. METHODS: In this study, we screened a large cohort of subjects with increased risk for diabetes and identified two subjects with impaired glucose tolerance carrying common, heterozygous, missense mutations in the PDX1 coding region leading to single amino acid exchanges (P33T, C18R) in its transactivation domain. We generated iPSCs from patients with heterozygous PDX1P33T/+, PDX1C18R/+ mutations and engineered isogenic cell lines carrying homozygous PDX1P33T/P33T, PDX1C18R/C18R mutations and a heterozygous PDX1 loss-of-function mutation (PDX1+/-). RESULTS: Using an in vitro ß-cell differentiation protocol, we demonstrated that both, heterozygous PDX1P33T/+, PDX1C18R/+ and homozygous PDX1P33T/P33T, PDX1C18R/C18R mutations impair ß-cell differentiation and function. Furthermore, PDX1+/- and PDX1P33T/P33T mutations reduced differentiation efficiency of pancreatic progenitors (PPs), due to downregulation of PDX1-bound genes, including transcription factors MNX1 and PDX1 as well as insulin resistance gene CES1. Additionally, both PDX1P33T/+ and PDX1P33T/P33T mutations in PPs reduced the expression of PDX1-bound genes including the long-noncoding RNA, MEG3 and the imprinted gene NNAT, both involved in insulin synthesis and secretion. CONCLUSIONS: Our results reveal mechanistic details of how common coding mutations in PDX1 impair human pancreatic endocrine lineage formation and ß-cell function and contribute to the predisposition for diabetes.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/genética , Proteínas de Homeodominio/genética , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Mutación Puntual , Transactivadores/genética , Adulto , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular , Femenino , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Células Secretoras de Insulina/citología , Mutación con Pérdida de Función , Masculino , Dominios Proteicos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Development ; 145(18)2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30126902

RESUMEN

During mouse pancreas organogenesis, endocrine cells are born from progenitors residing in an epithelial plexus niche. After a period in a lineage-primed Neurog3LO state, progenitors become endocrine committed via upregulation of Neurog3 We find that the Neurog3LO to Neurog3HI transition is associated with distinct stages of an epithelial egression process: narrowing the apical surface of the cell, basalward cell movement and eventual cell-rear detachment from the apical lumen surface to allow clustering as nascent islets under the basement membrane. Apical narrowing, basalward movement and Neurog3 transcriptional upregulation still occur without Neurog3 protein, suggesting that morphogenetic cues deployed within the plexus initiate endocrine commitment upstream or independently of Neurog3. Neurog3 is required for cell-rear detachment and complete endocrine-cell birth. The ROCK-nmMyoII pathway coordinates epithelial-cell morphogenesis and the progression through Neurog3-expressing states. NmMyoII is necessary for apical narrowing, basalward cell displacement and Neurog3 upregulation, but all three are limited by ROCK activity. We propose that ROCK-nmMyoII activity, Neurog3 gene-dose and Notch signaling integrate endocrine fate allocation with epithelial plexus growth and morphogenesis, representing a feedback control circuit that coordinates morphogenesis with lineage diversification in the endocrine-birth niche.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Endocrinas/citología , Dosificación de Gen/genética , Proteínas del Tejido Nervioso/genética , Organogénesis/genética , Páncreas/embriología , Receptores Notch/genética , Quinasas Asociadas a rho/genética , Animales , Diferenciación Celular/genética , Movimiento Celular , Ratones , Ratones Transgénicos , Páncreas/citología , Células Madre/citología , Activación Transcripcional/genética
13.
Mol Metab ; 9: 57-68, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29396371

RESUMEN

OBJECTIVE: Homozygous loss-of-function mutations in the gene coding for the homeobox transcription factor (TF) PDX1 leads to pancreatic agenesis, whereas heterozygous mutations can cause Maturity-Onset Diabetes of the Young 4 (MODY4). Although the function of Pdx1 is well studied in pre-clinical models during insulin-producing ß-cell development and homeostasis, it remains elusive how this TF controls human pancreas development by regulating a downstream transcriptional program. Also, comparative studies of PDX1 binding patterns in pancreatic progenitors and adult ß-cells have not been conducted so far. Furthermore, many studies reported the association between single nucleotide polymorphisms (SNPs) and T2DM, and it has been shown that islet enhancers are enriched in T2DM-associated SNPs. Whether regions, harboring T2DM-associated SNPs are PDX1 bound and active at the pancreatic progenitor stage has not been reported so far. METHODS: In this study, we have generated a novel induced pluripotent stem cell (iPSC) line that efficiently differentiates into human pancreatic progenitors (PPs). Furthermore, PDX1 and H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) was used to identify PDX1 transcriptional targets and active enhancer and promoter regions. To address potential differences in the function of PDX1 during development and adulthood, we compared PDX1 binding profiles from PPs and adult islets. Moreover, combining ChIP-seq and GWAS meta-analysis data we identified T2DM-associated SNPs in PDX1 binding sites and active chromatin regions. RESULTS: ChIP-seq for PDX1 revealed a total of 8088 PDX1-bound regions that map to 5664 genes in iPSC-derived PPs. The PDX1 target regions include important pancreatic TFs, such as PDX1 itself, RFX6, HNF1B, and MEIS1, which were activated during the differentiation process as revealed by the active chromatin mark H3K27ac and mRNA expression profiling, suggesting that auto-regulatory feedback regulation maintains PDX1 expression and initiates a pancreatic TF program. Remarkably, we identified several PDX1 target genes that have not been reported in the literature in human so far, including RFX3, required for ciliogenesis and endocrine differentiation in mouse, and the ligand of the Notch receptor DLL1, which is important for endocrine induction and tip-trunk patterning. The comparison of PDX1 profiles from PPs and adult human islets identified sets of stage-specific target genes, associated with early pancreas development and adult ß-cell function, respectively. Furthermore, we found an enrichment of T2DM-associated SNPs in active chromatin regions from iPSC-derived PPs. Two of these SNPs fall into PDX1 occupied sites that are located in the intronic regions of TCF7L2 and HNF1B. Both of these genes are key transcriptional regulators of endocrine induction and mutations in cis-regulatory regions predispose to diabetes. CONCLUSIONS: Our data provide stage-specific target genes of PDX1 during in vitro differentiation of stem cells into pancreatic progenitors that could be useful to identify pathways and molecular targets that predispose for diabetes. In addition, we show that T2DM-associated SNPs are enriched in active chromatin regions at the pancreatic progenitor stage, suggesting that the susceptibility to T2DM might originate from imperfect execution of a ß-cell developmental program.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodominio/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/metabolismo , Transactivadores/genética , Proteínas de Unión al Calcio , Diferenciación Celular , Células Cultivadas , Ensamble y Desensamble de Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Elementos de Facilitación Genéticos , Estudio de Asociación del Genoma Completo , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Transactivadores/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
14.
Cell Rep ; 21(3): 666-678, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045835

RESUMEN

Spinal interneurons are critical modulators of motor circuit function. In the dorsal spinal cord, a set of interneurons called GABApre presynaptically inhibits proprioceptive sensory afferent terminals, thus negatively regulating sensory-motor signaling. Although deficits in presynaptic inhibition have been inferred in human motor diseases, including dystonia, it remains unclear whether GABApre circuit components are altered in these conditions. Here, we use developmental timing to show that GABApre neurons are a late Ptf1a-expressing subclass and localize to the intermediate spinal cord. Using a microarray screen to identify genes expressed in this intermediate population, we find the kelch-like family member Klhl14, implicated in dystonia through its direct binding with torsion-dystonia-related protein Tor1a. Furthermore, in Tor1a mutant mice in which Klhl14 and Tor1a binding is disrupted, formation of GABApre sensory afferent synapses is impaired. Our findings suggest a potential contribution of GABApre neurons to the deficits in presynaptic inhibition observed in dystonia.


Asunto(s)
Distonía/genética , Neuronas GABAérgicas/patología , Predisposición Genética a la Enfermedad , Interneuronas/patología , Red Nerviosa/patología , Médula Espinal/patología , Animales , Biomarcadores/metabolismo , Distonía/patología , Distonía/fisiopatología , Masculino , Ratones Mutantes , Chaperonas Moleculares/genética , Mutación/genética , Red Nerviosa/fisiopatología , Terminales Presinápticos/patología , Propiocepción , Médula Espinal/fisiopatología , Factores de Transcripción/metabolismo
15.
Nat Commun ; 8(1): 605, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928395

RESUMEN

Spatiotemporal balancing of cellular proliferation and differentiation is crucial for postnatal tissue homoeostasis and organogenesis. During embryonic development, pancreatic progenitors simultaneously proliferate and differentiate into the endocrine, ductal and acinar lineages. Using in vivo clonal analysis in the founder population of the pancreas here we reveal highly heterogeneous contribution of single progenitors to organ formation. While some progenitors are bona fide multipotent and contribute progeny to all major pancreatic cell lineages, we also identify numerous unipotent endocrine and ducto-endocrine bipotent clones. Single-cell transcriptional profiling at E9.5 reveals that endocrine-committed cells are molecularly distinct, whereas multipotent and bipotent progenitors do not exhibit different expression profiles. Clone size and composition support a probabilistic model of cell fate allocation and in silico simulations predict a transient wave of acinar differentiation around E11.5, while endocrine differentiation is proportionally decreased. Increased proliferative capacity of outer progenitors is further proposed to impact clonal expansion.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Organogénesis , Páncreas/embriología , Células Acinares , Animales , Simulación por Computador , Perfilación de la Expresión Génica , Ratones , Análisis de la Célula Individual
16.
Genesis ; 55(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28772022

RESUMEN

During pancreas organogenesis, Neurog3HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO ). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI ) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2 -M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular , Células Madre Embrionarias/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Células Madre Embrionarias/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Islotes Pancreáticos/citología , Ratones , Proteínas del Tejido Nervioso/metabolismo
18.
Development ; 144(2): 248-257, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27993987

RESUMEN

The transcription factor Pdx1 is required for multiple aspects of pancreatic organogenesis. It remains unclear to what extent Pdx1 expression and function depend upon trans-activation through 5' conserved cis-regulatory regions and, in particular, whether the mammal-specific Area II (-2139 to -1958 bp) affects minor or major aspects of organogenesis. We show that Area II is a primary effector of endocrine-selective transcription in epithelial multipotent cells, nascent endocrine progenitors, and differentiating and mature ß cells in vivo Pdx1ΔAREAII/- mice exhibit a massive reduction in endocrine progenitor cells and progeny hormone-producing cells, indicating that Area II activity is fundamental to mounting an effective endocrine lineage-specification program within the multipotent cell population. Creating an Area II-deleted state within already specified Neurog3-expressing endocrine progenitor cells increased the proportion of glucagon+ α relative to insulin+ ß cells, associated with the transcriptional and epigenetic derepression of the α-cell-determining Arx gene in endocrine progenitors. There were also glucagon and insulin co-expressing cells, and ß cells that were incapable of maturation. Creating the Pdx1ΔAREAII state after cells entered an insulin-expressing stage led to immature and dysfunctional islet ß cells carrying abnormal chromatin marking in vital ß-cell-associated genes. Therefore, trans-regulatory integration through Area II mediates a surprisingly extensive range of progenitor and ß-cell-specific Pdx1 functions.


Asunto(s)
Diferenciación Celular/genética , Elementos de Facilitación Genéticos , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/embriología , Transactivadores/metabolismo , Animales , Sitios de Unión/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Insulina/citología , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Mamíferos/embriología , Mamíferos/genética , Ratones , Ratones Transgénicos , Organogénesis/genética , Especificidad de la Especie
19.
Mol Cell Biol ; 36(24): 3033-3047, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27697859

RESUMEN

Maintenance of cell type identity is crucial for health, yet little is known of the regulation that sustains the long-term stability of differentiated phenotypes. To investigate the roles that key transcriptional regulators play in adult differentiated cells, we examined the effects of depletion of the developmental master regulator PTF1A on the specialized phenotype of the adult pancreatic acinar cell in vivo Transcriptome sequencing and chromatin immunoprecipitation sequencing results showed that PTF1A maintains the expression of genes for all cellular processes dedicated to the production of the secretory digestive enzymes, a highly attuned surveillance of unfolded proteins, and a heightened unfolded protein response (UPR). Control by PTF1A is direct on target genes and indirect through a ten-member transcription factor network. Depletion of PTF1A causes an imbalance that overwhelms the UPR, induces cellular injury, and provokes acinar metaplasia. Compromised cellular identity occurs by derepression of characteristic stomach genes, some of which are also associated with pancreatic ductal cells. The loss of acinar cell homeostasis, differentiation, and identity is directly relevant to the pathologies of pancreatitis and pancreatic adenocarcinoma.


Asunto(s)
Células Acinares/citología , Perfilación de la Expresión Génica/métodos , Páncreas Exocrino/citología , Factores de Transcripción/genética , Transcripción Genética , Células Acinares/metabolismo , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Homeostasis , Ratones , Páncreas Exocrino/metabolismo , Desplegamiento Proteico , Análisis de Secuencia de ARN/métodos , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada
20.
Genes Dev ; 30(16): 1852-65, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27585590

RESUMEN

The current model for endocrine cell specification in the pancreas invokes high-level production of the transcription factor Neurogenin 3 (Neurog3) in Sox9(+) bipotent epithelial cells as the trigger for endocrine commitment, cell cycle exit, and rapid delamination toward proto-islet clusters. This model posits a transient Neurog3 expression state and short epithelial residence period. We show, however, that a Neurog3(TA.LO) cell population, defined as Neurog3 transcriptionally active and Sox9(+) and often containing nonimmunodetectable Neurog3 protein, has a relatively high mitotic index and prolonged epithelial residency. We propose that this endocrine-biased mitotic progenitor state is functionally separated from a pro-ductal pool and endows them with long-term capacity to make endocrine fate-directed progeny. A novel BAC transgenic Neurog3 reporter detected two types of mitotic behavior in Sox9(+) Neurog3(TA.LO) progenitors, associated with progenitor pool maintenance or derivation of endocrine-committed Neurog3(HI) cells, respectively. Moreover, limiting Neurog3 expression dramatically increased the proportional representation of Sox9(+) Neurog3(TA.LO) progenitors, with a doubling of its mitotic index relative to normal Neurog3 expression, suggesting that low Neurog3 expression is a defining feature of this cycling endocrine-biased state. We propose that Sox9(+) Neurog3(TA.LO) endocrine-biased progenitors feed production of Neurog3(HI) endocrine-committed cells during pancreas organogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Endocrinas/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular/genética , Ratones , Mitosis , Páncreas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...