Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.835
Filtrar
1.
BMC Genomics ; 25(1): 553, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831310

RESUMEN

Development of the human pancreas requires the precise temporal control of gene expression via epigenetic mechanisms and the binding of key transcription factors. We quantified genome-wide patterns of DNA methylation in human fetal pancreatic samples from donors aged 6 to 21 post-conception weeks. We found dramatic changes in DNA methylation across pancreas development, with > 21% of sites characterized as developmental differentially methylated positions (dDMPs) including many annotated to genes associated with monogenic diabetes. An analysis of DNA methylation in postnatal pancreas tissue showed that the dramatic temporal changes in DNA methylation occurring in the developing pancreas are largely limited to the prenatal period. Significant differences in DNA methylation were observed between males and females at a number of autosomal sites, with a small proportion of sites showing sex-specific DNA methylation trajectories across pancreas development. Pancreas dDMPs were not distributed equally across the genome and were depleted in regulatory domains characterized by open chromatin and the binding of known pancreatic development transcription factors. Finally, we compared our pancreas dDMPs to previous findings from the human brain, identifying evidence for tissue-specific developmental changes in DNA methylation. This study represents the first systematic exploration of DNA methylation patterns during human fetal pancreas development and confirms the prenatal period as a time of major epigenomic plasticity.


Asunto(s)
Metilación de ADN , Páncreas , Humanos , Páncreas/metabolismo , Páncreas/embriología , Femenino , Masculino , Regulación del Desarrollo de la Expresión Génica , Islas de CpG , Epigénesis Genética , Genoma Humano , Feto/metabolismo
2.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38731997

RESUMEN

Glucose, the primary energy substrate for fetal oxidative processes and growth, is transferred from maternal to fetal circulation down a concentration gradient by placental facilitative glucose transporters. In sheep, SLC2A1 and SLC2A3 are the primary transporters available in the placental epithelium, with SLC2A3 located on the maternal-facing apical trophoblast membrane and SLC2A1 located on the fetal-facing basolateral trophoblast membrane. We have previously reported that impaired placental SLC2A3 glucose transport resulted in smaller, hypoglycemic fetuses with reduced umbilical artery insulin and glucagon concentrations, in addition to diminished pancreas weights. These findings led us to subject RNA derived from SLC2A3-RNAi (RNA interference) and NTS-RNAi (non-targeting sequence) fetal pancreases to qPCR followed by transcriptomic analysis. We identified a total of 771 differentially expressed genes (DEGs). Upregulated pathways were associated with fat digestion and absorption, particularly fatty acid transport, lipid metabolism, and cholesterol biosynthesis, suggesting a potential switch in energetic substrates due to hypoglycemia. Pathways related to molecular transport and cell signaling in addition to pathways influencing growth and metabolism of the developing pancreas were also impacted. A few genes directly related to gluconeogenesis were also differentially expressed. Our results suggest that fetal hypoglycemia during the first half of gestation impacts fetal pancreas development and function that is not limited to ß cell activity.


Asunto(s)
Hipoglucemia , Páncreas , Placenta , Interferencia de ARN , Transcriptoma , Embarazo , Animales , Femenino , Placenta/metabolismo , Ovinos , Páncreas/metabolismo , Páncreas/embriología , Hipoglucemia/genética , Hipoglucemia/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Feto/metabolismo , Desarrollo Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Glucosa/metabolismo , Perfilación de la Expresión Génica
3.
Development ; 151(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38727565

RESUMEN

Proper embryonic development depends on the timely progression of a genetic program. One of the key mechanisms for achieving precise control of developmental timing is to use gene expression oscillations. In this Review, we examine how gene expression oscillations encode temporal information during vertebrate embryonic development by discussing the gene expression oscillations occurring during somitogenesis, neurogenesis, myogenesis and pancreas development. These oscillations play important but varied physiological functions in different contexts. Oscillations control the period of somite formation during somitogenesis, whereas they regulate the proliferation-to-differentiation switch of stem cells and progenitor cells during neurogenesis, myogenesis and pancreas development. We describe the similarities and differences of the expression pattern in space (i.e. whether oscillations are synchronous or asynchronous across neighboring cells) and in time (i.e. different time scales) of mammalian Hes/zebrafish Her genes and their targets in different tissues. We further summarize experimental evidence for the functional role of their oscillations. Finally, we discuss the outstanding questions for future research.


Asunto(s)
Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Somitos , Animales , Desarrollo Embrionario/genética , Humanos , Somitos/metabolismo , Somitos/embriología , Desarrollo de Músculos/genética , Neurogénesis/genética , Neurogénesis/fisiología , Páncreas/embriología , Páncreas/metabolismo , Diferenciación Celular/genética
4.
Diabetologia ; 67(6): 1066-1078, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38630142

RESUMEN

AIMS/HYPOTHESIS: Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS: We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS: We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION: This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Imagenología Tridimensional , Páncreas , Humanos , Páncreas/embriología , Páncreas/citología , Páncreas/metabolismo , Diferenciación Celular/fisiología , Femenino , Células Madre/citología , Células Madre/metabolismo , Embarazo , Insulina/metabolismo
5.
J Clin Ultrasound ; 52(5): 522-528, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38465703

RESUMEN

OBJECTIVE: To examine third-trimester sonographic imaging of the fetal pancreas in uncomplicated pregnancies and its association with pregnancy outcomes. METHODS: This was a prospective cohort study including 274 pregnant women. Uncomplicated pregnancies in the third trimester (28-40 weeks) were included in the study. Maternal chronic disease, pregnancy-related disorders such as hypertension, diabetes, cholestasis, smoking, and fetal abnormalities were determined as exclusion criteria. Sonographic fetal pancreatic measurement and echogenicity were evaluated in all participants. For intra-observer reliability, each participant's fetal pancreatic circumference was measured two times. The echogenicity of the pancreas was compared with the liver and ribs and classified as grade 1, 2, and 3. The pregnancy outcomes of all participants were obtained from the hospital digital registration system. RESULTS: The average fetal pancreatic circumference in the third trimester was 70.7 ± 0.6 mm (median, 70 [44-100.7]), with high intra-observer agreement (ICC 0.996 [0.995; 0.997]). A significant positive correlation was found between pancreatic circumference, body mass index (BMI), gestational age, and birth weight. Pancreatic measurements were significantly higher in composite adverse outcomes cases that included at least one of respiratory distress syndrome, hyperbilirubinemia, neonatal pneumonia, infection, and sepsis (p < 0.001). No relationship was found between pancreatic echogenicity and perinatal outcomes. CONCLUSION: Fetal pancreas size was positively correlated with gestational age, BMI, and birth weight, and increased fetal pancreas size was associated with composite adverse neonatal outcomes.


Asunto(s)
Páncreas , Resultado del Embarazo , Tercer Trimestre del Embarazo , Ultrasonografía Prenatal , Humanos , Embarazo , Femenino , Ultrasonografía Prenatal/métodos , Estudios Prospectivos , Adulto , Páncreas/diagnóstico por imagen , Páncreas/embriología , Reproducibilidad de los Resultados , Estudios de Cohortes
6.
Eur J Surg Oncol ; 50(6): 108272, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38552419

RESUMEN

BACKGROUND: Clinical progress in form of "total mesometrial resection" (TMMR) in cervical cancer and "total mesorectal excision" (TME) in rectal cancer can be traced to a paradigm-shift regarding the extent and range of resection. More significance is bestowed upon embryologically defined borders which define compartments, "morphogenetic units" and "cancer fields", that have to be addressed in order to avoid incomplete tumor resection. We want to transfer this rationale on the pancreas and define such borders for pancreatic compartments. MATERIAL AND METHODS: We used 26 unfixed body donors (16 male, 10 female) ranging in age from 64 to 98 years. Manual preparation consisted of performing the Cattell-Braasch maneuver to restore embryologic anatomy and define fascial remnants of the borders of the dorsal and ventral mesogastrium with focus on the pancreatic fusion fasciae and peripancreatic spaces. RESULTS: We tracked what used to be the dorsal and ventral mesogastrium and assigned their remnants to the bowel and pancreas. Following avascular embryologic fascial fusion planes along the mesogastria we could demonstrate peripancreatic spaces, which were sealed off from bordering surfaces of presumably different morphogenetic units and possible cancer fields. Reverting embryologic development also seemed possible within the pancreas, demonstrating the embryologic fusion plane between the ventral and dorsal pancreatic buds as two distinct compartments. CONCLUSIONS: Following pancreatic fusion fasciae by separating embryologic fusion planes enables to define the pancreatic compartments which might play a major role in applying the success of TMMR and TME on pancreatic resection and define pancreatic cancer fields.


Asunto(s)
Páncreas , Neoplasias Pancreáticas , Humanos , Femenino , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Persona de Mediana Edad , Anciano , Masculino , Anciano de 80 o más Años , Páncreas/embriología , Cadáver
7.
Am J Physiol Endocrinol Metab ; 322(2): E181-E196, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34957858

RESUMEN

Fetal hypoxemia decreases insulin and increases cortisol and norepinephrine concentrations and may restrict growth by decreasing glucose utilization and altering substrate oxidation. Specifically, we hypothesized that hypoxemia would decrease fetal glucose oxidation and increase lactate and pyruvate production. We tested this by measuring whole body glucose oxidation and lactate production, and molecular pathways in liver, muscle, adipose, and pancreas tissues of fetuses exposed to maternal hypoxemia for 9 days (HOX) compared with control fetal sheep (CON) in late gestation. Fetuses with more severe hypoxemia had lower whole body glucose oxidation rates, and HOX fetuses had increased lactate production from glucose. In muscle and adipose tissue, expression of the glucose transporter GLUT4 was decreased. In muscle, pyruvate kinase (PKM) and lactate dehydrogenase B (LDHB) expression was decreased. In adipose tissue, LDHA and lactate transporter (MCT1) expression was increased. In liver, there was decreased gene expression of PKLR and MPC2 and phosphorylation of PDH, and increased LDHA gene and LDH protein abundance. LDH activity, however, was decreased only in HOX skeletal muscle. There were no differences in basal insulin signaling across tissues, nor differences in pancreatic tissue insulin content, ß-cell area, or genes regulating ß-cell function. Collectively, these results demonstrate coordinated metabolic responses across tissues in the hypoxemic fetus that limit glucose oxidation and increase lactate and pyruvate production. These responses may be mediated by hypoxemia-induced endocrine responses including increased norepinephrine and cortisol, which inhibit pancreatic insulin secretion resulting in lower insulin concentrations and decreased stimulation of glucose utilization.NEW & NOTEWORTHY Hypoxemia lowered fetal glucose oxidation rates, based on severity of hypoxemia, and increased lactate production. This was supported by tissue-specific metabolic responses that may result from increased norepinephrine and cortisol concentrations, which decrease pancreatic insulin secretion and insulin concentrations and decrease glucose utilization. This highlights the vulnerability of metabolic pathways in the fetus and demonstrates that constrained glucose oxidation may represent an early event in response to sustained hypoxemia and fetal growth restriction.


Asunto(s)
Tejido Adiposo/metabolismo , Hipoxia Fetal/metabolismo , Feto/metabolismo , Glucosa/metabolismo , Ácido Láctico/biosíntesis , Hígado/metabolismo , Músculo Esquelético/metabolismo , Páncreas/metabolismo , Tejido Adiposo/embriología , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Insulina/metabolismo , Secreción de Insulina , Hígado/embriología , Masculino , Músculo Esquelético/embriología , Oxidación-Reducción , Páncreas/embriología , Embarazo , Ovinos
8.
Nat Commun ; 12(1): 6636, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789735

RESUMEN

FOXA pioneer transcription factors (TFs) associate with primed enhancers in endodermal organ precursors. Using a human stem cell model of pancreas differentiation, we here discover that only a subset of pancreatic enhancers is FOXA-primed, whereas the majority is unprimed and engages FOXA upon lineage induction. Primed enhancers are enriched for signal-dependent TF motifs and harbor abundant and strong FOXA motifs. Unprimed enhancers harbor fewer, more degenerate FOXA motifs, and FOXA recruitment to unprimed but not primed enhancers requires pancreatic TFs. Strengthening FOXA motifs at an unprimed enhancer near NKX6.1 renders FOXA recruitment pancreatic TF-independent, induces priming, and broadens the NKX6.1 expression domain. We make analogous observations about FOXA binding during hepatic and lung development. Our findings suggest a dual role for FOXA in endodermal organ development: first, FOXA facilitates signal-dependent lineage initiation via enhancer priming, and second, FOXA enforces organ cell type-specific gene expression via indirect recruitment by lineage-specific TFs.


Asunto(s)
Endodermo/embriología , Elementos de Facilitación Genéticos/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Sitios de Unión , Diferenciación Celular , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Hígado/embriología , Pulmón/embriología , Motivos de Nucleótidos , Especificidad de Órganos , Organogénesis , Páncreas/embriología , Transactivadores/genética
9.
Sci Rep ; 11(1): 22717, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34811400

RESUMEN

Retinoic acid (RA) is a key signal for the specification of the pancreas. Still, the gene regulatory cascade triggered by RA in the endoderm remains poorly characterized. In this study, we investigated this regulatory network in zebrafish by combining RNA-seq, RAR ChIP-seq and ATAC-seq assays. By analysing the effect of RA and of the RA receptor (RAR) inverse-agonist BMS493 on the transcriptome and on the chromatin accessibility of endodermal cells, we identified a large set of genes and regulatory regions regulated by RA signalling. RAR ChIP-seq further defined the direct RAR target genes in zebrafish, including hox genes as well as several pancreatic regulators like mnx1, insm1b, hnf1ba and gata6. Comparison of zebrafish and murine RAR ChIP-seq data highlighted the conserved direct target genes and revealed that some RAR sites are under strong evolutionary constraints. Among them, a novel highly conserved RAR-induced enhancer was identified downstream of the HoxB locus and driving expression in the nervous system and in the gut in a RA-dependent manner. Finally, ATAC-seq data unveiled the role of the RAR-direct targets Hnf1ba and Gata6 in opening chromatin at many regulatory loci upon RA treatment.


Asunto(s)
Genómica , Páncreas/efectos de los fármacos , Receptores de Ácido Retinoico/agonistas , Transcriptoma , Tretinoina/farmacología , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Páncreas/embriología , Páncreas/metabolismo , RNA-Seq , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Nat Med ; 27(11): 1928-1940, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663987

RESUMEN

Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Factor Nuclear 6 del Hepatocito/genética , Páncreas/embriología , Diferenciación Celular/genética , Anomalías Congénitas/genética , Retardo del Crecimiento Fetal/genética , Vesícula Biliar/anomalías , Proteína Homeobox Nkx-2.2/biosíntesis , Proteínas de Homeodominio/biosíntesis , Humanos , Lactante , Recién Nacido , Masculino , Herencia Multifactorial/genética , Organogénesis/genética , Páncreas/anomalías , Enfermedades Pancreáticas/congénito , Enfermedades Pancreáticas/genética , Células Madre Pluripotentes/citología , Transcripción Genética/genética
11.
Nature ; 597(7874): 87-91, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34433966

RESUMEN

Studies based on single cells have revealed vast cellular heterogeneity in stem cell and progenitor compartments, suggesting continuous differentiation trajectories with intermixing of cells at various states of lineage commitment and notable degrees of plasticity during organogenesis1-5. The hepato-pancreato-biliary organ system relies on a small endoderm progenitor compartment that gives rise to a variety of different adult tissues, including the liver, pancreas, gall bladder and extra-hepatic bile ducts6,7. Experimental manipulation of various developmental signals in the mouse embryo has underscored important cellular plasticity in this embryonic territory6. This is reflected in the existence of human genetic syndromes as well as congenital malformations featuring multi-organ phenotypes in liver, pancreas and gall bladder6. Nevertheless, the precise lineage hierarchy and succession of events leading to the segregation of an endoderm progenitor compartment into hepatic, biliary and pancreatic structures have not yet been established. Here we combine computational modelling approaches with genetic lineage tracing to accurately reconstruct the hepato-pancreato-biliary lineage tree. We show that a multipotent progenitor subpopulation persists in the pancreato-biliary organ rudiment, contributing cells not only to the pancreas and gall bladder but also to the liver. Moreover, using single-cell RNA sequencing and functional experiments we define a specialized niche that supports this subpopulation in a multipotent state for an extended time during development. Together these findings indicate sustained plasticity underlying hepato-pancreato-biliary development that might also explain the rapid expansion of the liver while attenuating pancreato-biliary growth.


Asunto(s)
Sistema Biliar/citología , Linaje de la Célula , Hígado/citología , Páncreas/citología , Nicho de Células Madre , Animales , Sistema Biliar/embriología , Sistema Biliar/metabolismo , Linaje de la Célula/genética , Rastreo Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Páncreas/embriología , Páncreas/metabolismo , RNA-Seq , Transducción de Señal , Análisis de la Célula Individual , Nicho de Células Madre/genética
12.
Stem Cell Reports ; 16(9): 2289-2304, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34450036

RESUMEN

Heterozygous mutations in HNF1B in humans result in a multisystem disorder, including pancreatic hypoplasia and diabetes mellitus. Here we used a well-controlled human induced pluripotent stem cell pancreatic differentiation model to elucidate the molecular mechanisms underlying HNF1B-associated diabetes. Our results show that lack of HNF1B blocks specification of pancreatic fate from the foregut progenitor (FP) stage, but HNF1B haploinsufficiency allows differentiation of multipotent pancreatic progenitor cells (MPCs) and insulin-secreting ß-like cells. We show that HNF1B haploinsufficiency impairs cell proliferation in FPs and MPCs. This could be attributed to impaired induction of key pancreatic developmental genes, including SOX11, ROBO2, and additional TEAD1 target genes whose function is associated with MPC self-renewal. In this work we uncover an exhaustive list of potential HNF1B gene targets during human pancreas organogenesis whose downregulation might underlie HNF1B-associated diabetes onset in humans, thus providing an important resource to understand the pathogenesis of this disease.


Asunto(s)
Diferenciación Celular/genética , Factor Nuclear 1-beta del Hepatocito/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Organogénesis/genética , Páncreas/embriología , Páncreas/metabolismo , Biomarcadores , Sistemas CRISPR-Cas , Linaje de la Célula/genética , Diabetes Mellitus/etiología , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Edición Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia , Factor Nuclear 1-beta del Hepatocito/metabolismo , Humanos , Inmunofenotipificación , Células Secretoras de Insulina/metabolismo , Transducción de Señal
13.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201511

RESUMEN

Diabetes is a metabolic disease that involves the death or dysfunction of the insulin-secreting ß cells in the pancreas. Consequently, most diabetes research is aimed at understanding the molecular and cellular bases of pancreatic development, islet formation, ß-cell survival, and insulin secretion. Complex interactions of signaling pathways and transcription factor networks regulate the specification, growth, and differentiation of cell types in the developing pancreas. Many of the same regulators continue to modulate gene expression and cell fate of the adult pancreas. The transcription factor NEUROD1 is essential for the maturation of ß cells and the expansion of the pancreatic islet cell mass. Mutations of the Neurod1 gene cause diabetes in humans and mice. However, the different aspects of the requirement of NEUROD1 for pancreas development are not fully understood. In this study, we investigated the role of NEUROD1 during the primary and secondary transitions of mouse pancreas development. We determined that the elimination of Neurod1 impairs the expression of key transcription factors for α- and ß-cell differentiation, ß-cell proliferation, insulin production, and islets of Langerhans formation. These findings demonstrate that the Neurod1 deletion altered the properties of α and ß endocrine cells, resulting in severe neonatal diabetes, and thus, NEUROD1 is required for proper activation of the transcriptional network and differentiation of functional α and ß cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Islotes Pancreáticos/citología , Páncreas/citología , Páncreas/embriología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Diabetes Mellitus/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
Nat Commun ; 12(1): 4458, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294685

RESUMEN

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.


Asunto(s)
Insulina/biosíntesis , Células Secretoras de Polipéptido Pancreático/metabolismo , Polipéptido Pancreático/metabolismo , Precursores de Proteínas/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal , Linaje de la Célula/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Células Secretoras de Insulina/clasificación , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Páncreas/citología , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Polipéptido Pancreático/deficiencia , Polipéptido Pancreático/genética , Células Secretoras de Polipéptido Pancreático/clasificación , Células Secretoras de Polipéptido Pancreático/citología , Embarazo , RNA-Seq
15.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34299221

RESUMEN

During the early developmental stages of grass snakes, within the differentiating pancreas, cords of endocrine cells are formed. They differentiate into agglomerates of large islets flanked throughout subsequent developmental stages by small groups of endocrine cells forming islets. The islets are located within the cephalic part of the dorsal pancreas. At the end of the embryonic period, the pancreatic islet agglomerates branch off, and as a result of their remodeling, surround the splenic "bulb". The stage of pancreatic endocrine ring formation is the first step in formation of intrasplenic islets characteristics for the adult specimens of the grass snake. The arrangement of endocrine cells within islets changes during pancreas differentiation. Initially, the core of islets formed from B and D cells is surrounded by a cluster of A cells. Subsequently, A, B, and D endocrine cells are mixed throughout the islets. Before grass snake hatching, A and B endocrine cells are intermingled within the islets, but D cells are arranged centrally. Moreover, the pancreatic polypeptide (PP) cells are not found within the embryonic pancreas of the grass snake. Variation in the proportions of different cell types, depending on the part of the pancreas, may affect the islet function-a higher proportion of glucagon cells is beneficial for insulin secretion.


Asunto(s)
Colubridae/embriología , Islotes Pancreáticos/embriología , Páncreas/embriología , Animales , Diferenciación Celular , Colubridae/metabolismo , Células Endocrinas/metabolismo , Células Endocrinas/fisiología , Sistema Endocrino/metabolismo , Imagenología Tridimensional , Insulina/metabolismo , Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/inmunología , Páncreas/anatomía & histología , Páncreas/inmunología
16.
Sci Rep ; 11(1): 14519, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34267243

RESUMEN

Tight junction complexes are involved in the establishment and maintenance of cell polarity and the regulation of signalling pathways, controlling biological processes such as cell differentiation and cell proliferation. MarvelD3 is a tight junction protein expressed in adult epithelial and endothelial cells. In Xenopus laevis, MarvelD3 morphants present differentiation defects of several ectodermal derivatives. In vitro experiments further revealed that MarvelD3 couples tight junctions to the MEKK1-JNK pathway to regulate cell behaviour and survival. In this work, we found that MarvelD3 is expressed from early developmental stages in the exocrine and endocrine compartments of the pancreas, as well as in endothelial cells of this organ. We thoroughly characterized MarvelD3 expression pattern in developing pancreas and evaluated its function by genetic ablation. Surprisingly, inactivation of MarvelD3 in mice did not alter development and differentiation of the pancreatic tissue. Moreover, tight junction formation and organization, cell polarization, and activity of the JNK-pathway were not impacted by the deletion of MarvelD3.


Asunto(s)
Proteínas con Dominio MARVEL/genética , Páncreas/embriología , Páncreas/fisiología , Proteínas de Uniones Estrechas/genética , Animales , Sistemas CRISPR-Cas , Diferenciación Celular/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Sistema de Señalización de MAP Quinasas/genética , Proteínas con Dominio MARVEL/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/citología , Glándulas Salivales/fisiología , Análisis Espacio-Temporal , Proteínas de Uniones Estrechas/metabolismo
17.
J Endocrinol ; 250(3): R23-R35, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34196608

RESUMEN

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.


Asunto(s)
Diabetes Mellitus/genética , Células Secretoras de Insulina/fisiología , Insulina/historia , Animales , Diferenciación Celular/genética , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/historia , Predisposición Genética a la Enfermedad , Genómica/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Insulina/genética , Insulina/uso terapéutico , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo
18.
Sci Rep ; 11(1): 15475, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34326390

RESUMEN

The apelin receptor (Aplnr) and its ligands, Apelin and Apela, contribute to metabolic control. The insulin resistance associated with pregnancy is accommodated by an expansion of pancreatic ß-cell mass (BCM) and increased insulin secretion, involving the proliferation of insulin-expressing, glucose transporter 2-low (Ins+Glut2LO) progenitor cells. We examined changes in the apelinergic system during normal mouse pregnancy and in pregnancies complicated by glucose intolerance with reduced BCM. Expression of Aplnr, Apelin and Apela was quantified in Ins+Glut2LO cells isolated from mouse pancreata and found to be significantly higher than in mature ß-cells by DNA microarray and qPCR. Apelin was localized to most ß-cells by immunohistochemistry although Aplnr was predominantly associated with Ins+Glut2LO cells. Aplnr-staining cells increased three- to four-fold during pregnancy being maximal at gestational days (GD) 9-12 but were significantly reduced in glucose intolerant mice. Apelin-13 increased ß-cell proliferation in isolated mouse islets and INS1E cells, but not glucose-stimulated insulin secretion. Glucose intolerant pregnant mice had significantly elevated serum Apelin levels at GD 9 associated with an increased presence of placental IL-6. Placental expression of the apelinergic axis remained unaltered, however. Results show that the apelinergic system is highly expressed in pancreatic ß-cell progenitors and may contribute to ß-cell proliferation in pregnancy.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Páncreas/embriología , Preñez , Animales , Apelina/metabolismo , Receptores de Apelina/metabolismo , Proliferación Celular , Separación Celular , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Intolerancia a la Glucosa , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Placenta/metabolismo , Embarazo
19.
Front Endocrinol (Lausanne) ; 12: 648284, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079523

RESUMEN

Diabetes, as one of the major diseases in industrial countries, affects over 350 million people worldwide. Type 1 (T1D) and type 2 diabetes (T2D) are the most common forms with both types having invariable genetic influence. It is accepted that a subset of all diabetes patients, generally estimated to account for 1-2% of all diabetic cases, is attributed to mutations in single genes. As only a subset of these genes has been identified and fully characterized, there is a dramatic need to understand the pathophysiological impact of genetic determinants on ß-cell function and pancreatic development but also on cell replacement therapies. Pluripotent stem cells differentiated along the pancreatic lineage provide a valuable research platform to study such genes. This review summarizes current perspectives in applying this platform to study monogenic diabetes variants.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Mutación , Células Madre Pluripotentes/citología , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Epigénesis Genética , Edición Génica , Variación Genética , Heterocigoto , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Páncreas/embriología , Páncreas/patología , Fenotipo , Regeneración
20.
Curr Opin Genet Dev ; 70: 32-39, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34062490

RESUMEN

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.


Asunto(s)
Reprogramación Celular/fisiología , Páncreas/citología , Animales , Linaje de la Célula , Plasticidad de la Célula , Técnicas de Reprogramación Celular/métodos , Regulación de la Expresión Génica , Humanos , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA