Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(26): e2201267119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35733248

RESUMEN

Delineating gene regulatory networks that orchestrate cell-type specification is a continuing challenge for developmental biologists. Single-cell analyses offer opportunities to address these challenges and accelerate discovery of rare cell lineage relationships and mechanisms underlying hierarchical lineage decisions. Here, we describe the molecular analysis of mouse pancreatic endocrine cell differentiation using single-cell transcriptomics, chromatin accessibility assays coupled to genetic labeling, and cytometry-based cell purification. We uncover transcription factor networks that delineate ß-, α-, and δ-cell lineages. Through genomic footprint analysis, we identify transcription factor-regulatory DNA interactions governing pancreatic cell development at unprecedented resolution. Our analysis suggests that the transcription factor Neurog3 may act as a pioneer transcription factor to specify the pancreatic endocrine lineage. These findings could improve protocols to generate replacement endocrine cells from renewable sources, like stem cells, for diabetes therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cromatina , Islotes Pancreáticos , Proteínas del Tejido Nervioso , Transcriptoma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Cromatina/genética , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis de la Célula Individual
2.
Development ; 147(6)2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32108026

RESUMEN

Reliance on rodents for understanding pancreatic genetics, development and islet function could limit progress in developing interventions for human diseases such as diabetes mellitus. Similarities of pancreas morphology and function suggest that porcine and human pancreas developmental biology may have useful homologies. However, little is known about pig pancreas development. To fill this knowledge gap, we investigated fetal and neonatal pig pancreas at multiple, crucial developmental stages using modern experimental approaches. Purification of islet ß-, α- and δ-cells followed by transcriptome analysis (RNA-seq) and immunohistology identified cell- and stage-specific regulation, and revealed that pig and human islet cells share characteristic features that are not observed in mice. Morphometric analysis also revealed endocrine cell allocation and architectural similarities between pig and human islets. Our analysis unveiled scores of signaling pathways linked to native islet ß-cell functional maturation, including evidence of fetal α-cell GLP-1 production and signaling to ß-cells. Thus, the findings and resources detailed here show how pig pancreatic islet studies complement other systems for understanding the developmental programs that generate functional islet cells, and that are relevant to human pancreatic diseases.


Asunto(s)
Diferenciación Celular/genética , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/embriología , Islotes Pancreáticos/crecimiento & desarrollo , Porcinos , Animales , Animales Recién Nacidos , Células Cultivadas , Embrión de Mamíferos , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/fisiología , Humanos , Islotes Pancreáticos/citología , Ratones , Organogénesis/genética , Embarazo , Porcinos/embriología , Porcinos/genética , Porcinos/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
3.
Development ; 144(20): 3744-3754, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28893946

RESUMEN

The islets of Langerhans are endocrine organs characteristically dispersed throughout the pancreas. During development, endocrine progenitors delaminate, migrate radially and cluster to form islets. Despite the distinctive distribution of islets, spatially localized signals that control islet morphogenesis have not been discovered. Here, we identify a radial signaling axis that instructs developing islet cells to disperse throughout the pancreas. A screen of pancreatic extracellular signals identified factors that stimulated islet cell development. These included semaphorin 3a, a guidance cue in neural development without known functions in the pancreas. In the fetal pancreas, peripheral mesenchymal cells expressed Sema3a, while central nascent islet cells produced the semaphorin receptor neuropilin 2 (Nrp2). Nrp2 mutant islet cells developed in proper numbers, but had defects in migration and were unresponsive to purified Sema3a. Mutant Nrp2 islets aggregated centrally and failed to disperse radially. Thus, Sema3a-Nrp2 signaling along an unrecognized pancreatic developmental axis constitutes a chemoattractant system essential for generating the hallmark morphogenetic properties of pancreatic islets. Unexpectedly, Sema3a- and Nrp2-mediated control of islet morphogenesis is strikingly homologous to mechanisms that regulate radial neuronal migration and cortical lamination in the developing mammalian brain.


Asunto(s)
Islotes Pancreáticos/citología , Neuropilina-2/metabolismo , Semaforina-3A/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Factores Quimiotácticos/química , Regulación del Desarrollo de la Expresión Génica , Humanos , Ligandos , Ratones , Ratones Noqueados , Morfogénesis , Mutación , Neuronas/metabolismo , Neuropilina-2/genética , Páncreas/citología , Semaforina-3A/genética , Transducción de Señal
4.
STAR Protoc ; 5(3): 103107, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38963758

RESUMEN

Electroporation is a technique to introduce DNA constructs into cells using electric current. Here, we present a protocol to electroporate DNA plasmids into Ciona robusta embryos at the 1-cell stage. We describe steps for setting up and conducting electroporation. We then detail procedures for collecting, fixing, and mounting embryos and counting expression. This protocol can be used to study the expression of enhancers via reporter assays, manipulating cells using genes or modified genes such as dominant negatives, and genome editing. For complete details on the use and execution of this protocol, please refer to Song, et al.1.

5.
Diabetes ; 73(3): 448-460, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38064570

RESUMEN

Mutations in the gene encoding the transcription factor regulatory factor X-box binding 6 (RFX6) are associated with human diabetes. Within pancreatic islets, RFX6 expression is most abundant in islet α-cells, and α-cell RFX6 expression is altered in diabetes. However, the roles of RFX6 in regulating gene expression, glucagon output, and other crucial human adult α-cell functions are not yet understood. We developed a method for selective genetic targeting of human α-cells and assessed RFX6-dependent α-cell function. RFX6 suppression with RNA interference led to impaired α-cell exocytosis and dysregulated glucagon secretion in vitro and in vivo. By contrast, these phenotypes were not observed with RFX6 suppression across all islet cells. Transcriptomics in α-cells revealed RFX6-dependent expression of genes governing nutrient sensing, hormone processing, and secretion, with some of these exclusively expressed in human α-cells. Mapping of RFX6 DNA-binding sites in primary human islet cells identified a subset of direct RFX6 target genes. Together, these data unveil RFX6-dependent genetic targets and mechanisms crucial for regulating adult human α-cell function.


Asunto(s)
Diabetes Mellitus , Islotes Pancreáticos , Humanos , Glucagón/metabolismo , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Islotes Pancreáticos/metabolismo , Diabetes Mellitus/metabolismo , Expresión Génica , Insulina/metabolismo
6.
Cell Rep ; 42(2): 112052, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36729834

RESUMEN

The notochord is a defining feature of all chordates. The transcription factors Zic and ETS regulate enhancer activity within the notochord. We conduct high-throughput screens of genomic elements within developing Ciona embryos to understand how Zic and ETS sites encode notochord activity. Our screen discovers an enhancer located near Lama, a gene critical for notochord development. Reversing the orientation of an ETS site within this enhancer abolishes expression, indicating that enhancer grammar is critical for notochord activity. Similarly organized clusters of Zic and ETS sites occur within mouse and human Lama1 introns. Within a Brachyury (Bra) enhancer, FoxA and Bra, in combination with Zic and ETS binding sites, are necessary and sufficient for notochord expression. This binding site logic also occurs within other Ciona and vertebrate Bra enhancers. Collectively, this study uncovers the importance of grammar within notochord enhancers and discovers signatures of enhancer logic and grammar conserved across chordates.


Asunto(s)
Ciona intestinalis , Notocorda , Animales , Humanos , Ratones , Ciona intestinalis/genética , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Regulación del Desarrollo de la Expresión Génica , Elementos de Facilitación Genéticos/genética
7.
Nat Commun ; 12(1): 2397, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893274

RESUMEN

Gene targeting studies in primary human islets could advance our understanding of mechanisms driving diabetes pathogenesis. Here, we demonstrate successful genome editing in primary human islets using clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9). CRISPR-based targeting efficiently mutated protein-coding exons, resulting in acute loss of islet ß-cell regulators, like the transcription factor PDX1 and the KATP channel subunit KIR6.2, accompanied by impaired ß-cell regulation and function. CRISPR targeting of non-coding DNA harboring type 2 diabetes (T2D) risk variants revealed changes in ABCC8, SIX2 and SIX3 expression, and impaired ß-cell function, thereby linking regulatory elements in these target genes to T2D genetic susceptibility. Advances here establish a paradigm for genetic studies in human islet cells, and reveal regulatory and genetic mechanisms linking non-coding variants to human diabetes risk.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Modelos Genéticos , Secuencia de Bases , Diabetes Mellitus Tipo 2/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Canales de Potasio de Rectificación Interna/genética , Transactivadores/genética
8.
Nat Metab ; 2(6): 547-557, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32694729

RESUMEN

Little is known about regulated glucagon secretion by human islet α-cells compared to insulin secretion from ß-cells, despite conclusive evidence of dysfunction in both cell types in diabetes mellitus. Distinct insulins in humans and mice permit in vivo studies of human ß-cell regulation after human islet transplantation in immunocompromised mice, whereas identical glucagon sequences prevent analogous in vivo measures of glucagon output from human α-cells. Here, we use CRISPR-Cas9 editing to remove glucagon codons 2-29 in immunocompromised NSG mice, preserving the production of other proglucagon-derived hormones. Glucagon knockout NSG (GKO-NSG) mice have metabolic, liver and pancreatic phenotypes associated with glucagon-signalling deficits that revert after transplantation of human islets from non-diabetic donors. Glucagon hypersecretion by transplanted islets from donors with type 2 diabetes revealed islet-intrinsic defects. We suggest that GKO-NSG mice provide an unprecedented resource to investigate human α-cell regulation in vivo.


Asunto(s)
Glucagón/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Adulto , Animales , Sistemas CRISPR-Cas , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucagón/genética , Células Secretoras de Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA