Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
J Anat ; 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38581181

ABSTRACT

This study investigated the pancreas differentiation of two species of gekkotan families-the mourning gecko Lepidodactylus lugubris (Gekkonidae) and the leopard gecko Eublepharis macularius (Eublepharidae)-based on two-dimensional (2D) histological samples and three-dimensional (3D) reconstructions of the position of the pancreatic buds and the surrounding organs. The results showed that at the moment of egg laying, the pancreas of L. lugubris is composed of three distinct primordia: one dorsal and two ventral. The dorsal primordium differentiates earlier than either ventral primordium. The right ventral primordium is more prominent and distinctive, starting to form earlier than the left one. Moreover, at this time, the pancreas of the leopard gecko is composed of the dorsal and right ventral primordium and the duct of the left ventral primordium. It means that the leopard gecko's left primordium is a transitional structure. These results indicate that the early development of the gekkotan pancreas is species specific. The pancreatic buds of the leopard and mourning gecko initially enter the duodenum by separate outlets, similar to the pancreas of other vertebrates. The pancreatic buds (3 of the mourning gecko and 2 of the leopard gecko) fuse quickly and form an embryonic pancreas. After that, the structure of this organ changes. After fusion, the pancreas of both gekkotans comprises four parts: the head of the pancreas (central region) and three lobes: upper, splenic, and lower. This organ develops gradually and is very well distinguished at hatching time. In both gekkotan species, cystic, hepatic, and pancreatic ducts enter the duodenum within the papilla. During gekkotan pancreas differentiation, the connection between the common bile duct and the dorsal pancreatic duct is associated with intestinal rotation, similar to other vertebrates.

2.
Cell Rep ; 43(2): 113703, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38265933

ABSTRACT

Pancreas development is tightly controlled by multilayer mechanisms. Despite years of effort, large gaps remain in understanding how histone modifications coordinate pancreas development. SETD2, a predominant histone methyltransferase of H3K36me3, plays a key role in embryonic stem cell differentiation, whose role in organogenesis remains elusive. Here, by combination of cleavage under targets and tagmentation (CUT&Tag), assay for transposase-accessible chromatin using sequencing (ATAC-seq), and bulk RNA sequencing, we show a dramatic increase in the H3K36me3 level from the secondary transition phase and decipher the related transcriptional alteration. Using single-cell RNA sequencing, we define that pancreatic deletion of Setd2 results in abnormalities in both exocrine and endocrine lineages: hyperproliferative tip progenitor cells lead to abnormal differentiation; Ngn3+ endocrine progenitors decline due to the downregulation of Nkx2.2, leading to insufficient endocrine development. Thus, these data identify SETD2 as a crucial player in embryonic pancreas development, providing a clue to understanding the dysregulation of histone modifications in pancreatic disorders.


Subject(s)
Chromatin , Pancreas , Animals , Mice , Cell Differentiation , Histone-Lysine N-Methyltransferase/genetics , Organogenesis/genetics
3.
Dev Biol ; 503: 43-52, 2023 11.
Article in English | MEDLINE | ID: mdl-37597605

ABSTRACT

Transmembrane p24 trafficking protein 10 (TMED10) is a conserved vesicle trafficking protein. It is dysregulated in Alzheimer disease and plays a pivotal role in the pathogenesis of Alzheimer disease. In addition to the brain, TMED10 is highly expressed in the exocrine pancreas; however, its biological functions and underlying mechanisms remain largely unknown. We studied reduced Tmed10 in zebrafish embryos by morpholino oligonucleotide knockdown and CRISPR-Cas9 mutagenesis. Tmed10-deficient embryos showed extensive loss of acinar mass and impaired acinar differentiation. TMED10 has been reported to have an inhibitory effect on γ-secretase. As one of the substrates of γ-secretase, membrane-bound ß-catenin was significantly reduced in Tmed10-deficient embryos. Increased γ-secretase activity in wild-type embryos resulted in a phenotype similar to that of tmed10 mutants. And the mutant phenotype could be rescued by treatment with the γ-secretase inhibitor, N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester (DAPT). In addition, the reduced membrane-bound ß-catenin was accompanied with up-regulated ß-catenin target genes in Tmed10-deficient embryos. Overexpression of ß-catenin signaling inhibitor Dickkopf-1 (DKK-1) could rescue the exocrine pancreas defects. Taken together, our study reveals that Tmed10 regulates exocrine pancreatic differentiation through γ-secretase. Reduced membrane-bound ß-catenin, accompanied with hyperactivation of ß-catenin signaling, is an important cause of exocrine pancreas defects in Tmed10-deficient embryos. Our study reaffirms the importance of appropriate ß-catenin signaling in exocrine pancreas development. These findings may provide a theoretical basis for the development of treatment strategies for TMED10-related diseases.


Subject(s)
Alzheimer Disease , Nucleocytoplasmic Transport Proteins , Pancreas, Exocrine , Animals , Amyloid Precursor Protein Secretases/genetics , beta Catenin/genetics , Larva , Pancreas, Exocrine/embryology , Pancreas, Exocrine/metabolism , Zebrafish/genetics , Nucleocytoplasmic Transport Proteins/metabolism
4.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36897571

ABSTRACT

Hormone secretion from pancreatic islets is essential for glucose homeostasis, and loss or dysfunction of islet cells is a hallmark of type 2 diabetes. Maf transcription factors are crucial for establishing and maintaining adult endocrine cell function. However, during pancreas development, MafB is not only expressed in insulin- and glucagon-producing cells, but also in Neurog3+ endocrine progenitor cells, suggesting additional functions in cell differentiation and islet formation. Here, we report that MafB deficiency impairs ß cell clustering and islet formation, but also coincides with loss of neurotransmitter and axon guidance receptor gene expression. Moreover, the observed loss of nicotinic receptor gene expression in human and mouse ß cells implied that signaling through these receptors contributes to islet cell migration/formation. Inhibition of nicotinic receptor activity resulted in reduced ß cell migration towards autonomic nerves and impaired ß cell clustering. These findings highlight a novel function of MafB in controlling neuronal-directed signaling events required for islet formation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Adult , Animals , Humans , Glucagon/genetics , Glucagon/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Pancreas/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism
5.
Cell Biosci ; 13(1): 53, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36899442

ABSTRACT

BACKGROUND: Glucose homeostasis is dependent on functional pancreatic α and ß cells. The mechanisms underlying the generation and maturation of these endocrine cells remain unclear. RESULTS: We unravel the molecular mode of action of ISL1 in controlling α cell fate and the formation of functional ß cells in the pancreas. By combining transgenic mouse models, transcriptomic and epigenomic profiling, we uncover that elimination of Isl1 results in a diabetic phenotype with a complete loss of α cells, disrupted pancreatic islet architecture, downregulation of key ß-cell regulators and maturation markers of ß cells, and an enrichment in an intermediate endocrine progenitor transcriptomic profile. CONCLUSIONS: Mechanistically, apart from the altered transcriptome of pancreatic endocrine cells, Isl1 elimination results in altered silencing H3K27me3 histone modifications in the promoter regions of genes that are essential for endocrine cell differentiation. Our results thus show that ISL1 transcriptionally and epigenetically controls α cell fate competence, and ß cell maturation, suggesting that ISL1 is a critical component for generating functional α and ß cells.

6.
Bioessays ; 45(4): e2200186, 2023 04.
Article in English | MEDLINE | ID: mdl-36871153

ABSTRACT

Unraveling molecular and functional heterogeneity of niche cells within the developing endoderm could resolve mechanisms of tissue formation and maturation. Here, we discuss current unknowns in molecular mechanisms underlying key developmental events in pancreatic islet and intestinal epithelial formation. Recent breakthroughs in single-cell and spatial transcriptomics, paralleled with functional studies in vitro, reveal that specialized mesenchymal subtypes drive the formation and maturation of pancreatic endocrine cells and islets via local interactions with epithelium, neurons, and microvessels. Analogous to this, distinct intestinal niche cells regulate both epithelial development and homeostasis throughout life. We propose how this knowledge can be used to progress research in the human context using pluripotent stem cell-derived multilineage organoids. Overall, understanding the interactions between the multitude of microenvironmental cells and how they drive tissue development and function could help us make more therapeutically relevant in vitro models.


Subject(s)
Endoderm , Pancreas , Humans , Cell Differentiation/physiology , Homeostasis , Intestines
7.
J Biol Chem ; 299(2): 102878, 2023 02.
Article in English | MEDLINE | ID: mdl-36623733

ABSTRACT

Deletion of O-GlcNAc transferase (Ogt) in pancreatic epithelial progenitor cells results in pancreatic hypoplasia at birth, partly due to increased apoptosis during embryonic development. Constitutive loss of Ogt in ß-cells results in increased ER stress and apoptosis, and in the Ogt-deficient pancreas, transcriptomic data previously revealed both tumor suppressor protein p53 and pancreatic duodenal homeobox 1 (Pdx1), key cell survival proteins in the developing pancreas, as upstream regulators of differentially expressed genes. However, the specific roles of these genes in pancreatic hypoplasia are unclear. In this study, we explored the independent roles of p53, ER stress protein CHOP, and Pdx1 in pancreas development and their use in the functional rescue of pancreatic hypoplasia in the context of Ogt loss. Using in vivo genetic manipulation and morphometric analysis, we show that Ogt plays a key regulatory role in pancreas development. Heterozygous, but not homozygous, loss of pancreatic p53 afforded a partial rescue of ß-cell, α-cell, and exocrine cell masses, while whole body loss of CHOP afforded a partial rescue in pancreas weight and a full rescue in exocrine cell mass. However, neither was sufficient to fully mitigate pancreatic hypoplasia at birth in the Ogt-deficient pancreas. Furthermore, overexpression of Pdx1 in the pancreatic epithelium resulted in partial rescues in pancreas weight and ß-cell mass in the Ogt loss background. These findings highlight the requirement of Ogt in pancreas development by targeting multiple proteins such as transcription factor Pdx1 and p53 in the developing pancreas.


Subject(s)
Gene Expression , Glucagon-Secreting Cells , Pancreatic Diseases , Tumor Suppressor Protein p53 , Animals , Mice , Glucagon-Secreting Cells/metabolism , Pancreas, Exocrine/metabolism , Tumor Suppressor Protein p53/genetics , Gene Expression/genetics , Pancreatic Diseases/genetics , Pancreatic Diseases/physiopathology
8.
Animals (Basel) ; 13(2)2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36670833

ABSTRACT

In this study, we used a combination of animal nutrition and nontargeted metabolomics to investigate the effects of feeding different sources forages rations on the morphology and function of the pancreas in preweaning yak calves, providing theoretical guidance and important references for the healthy and high-quality rearing of yak calves. At 45 days old, 21 yak calf males were divided into OP, AP, and AOP groups, with seven animals in each group, which were fed with oat hay, alfalfa hay, and mixed oat and alfalfa hay, respectively. Five calves from each group were selected randomly to slaughter after a pretest period of 21 days and the official period of 120 days, when the average daily feed intake reached 1 kg. During the test, the growth and pancreas weight of yak calves were recorded, and the morphology and function of the pancreas tissues were determined using tissue sectioning methods, enzyme-linked immunosorbent assay (ELISA) tests, and nontargeted metabolomics strategies. The results showed that the body weight and pancreatic organ index of yak calves in the AOP group were significantly higher than those of the AP and OP groups. Compared to the AP and OP groups, the AOP group had considerably lower ratios of the area of the pancreatic endocrine component and overall percentage of that section of the organ, and the AOP group increased pancreatic amylase activity and a higher concentration of growth inhibitor. The AP group had significantly higher levels of the differential metabolites L-ascorbic acid, spermidine, spermine, and dopaquinone in the glutathione, ß-alanine, and tyrosine metabolic pathways than the OP group. The AOP group had significantly lower levels of the differential metabolites spermine and phenylacetylglycine in the glutathione and phenylalanine metabolic pathways than the AP group. In summary, compared to feeding oat or alfalfa hay alone, combined feeding oat hay and alfalfa hay is more beneficial to promote the morphological and functional development of the pancreas in preweaning yak calves, so as to enhance the digestion and absorption of nutrients in the diet and maintain the positive regulation of blood glucose levels. This provides an important basis for the optimized forage supply of healthy and high-quality rearing in preweaning yak calves.

9.
Growth Factors ; 41(1): 8-19, 2023 02.
Article in English | MEDLINE | ID: mdl-36373834

ABSTRACT

Retinoic acid (RA) is essential for gut endoderm development and has been extensively used for in vitro pancreatic differentiation from human pluripotent stem cells. However, the gene regulatory network triggered by RA signaling remains poorly addressed. Also, whether RA signals control histone modifiers such as the Polycomb group proteins during pancreatic specification remains to be explored. Here, we assess the role of RA on pancreas-specific genes during the differentiation of human embryonic stem cells (hESCs). We demonstrate that RA helps cells exit the definitive endoderm stage and proceed toward a pancreatic fate. Inhibition of the RA pathway using the pharmacological inhibitor LE135 impairs the induction of pancreatic endoderm (PE) markers FOXA2, HNF4α, HNF1ß, HHEX, and PDX1. We further determine that RA signals alter the expression of epigenetic-associated genes BMI1 and RING1B in the hESC-derived pancreatic progenitors. These findings broaden our understanding of the mechanisms that drive early PE specification.


Subject(s)
Human Embryonic Stem Cells , Humans , Pancreas , Signal Transduction , Cell Differentiation , Homeodomain Proteins/genetics , Tretinoin/pharmacology
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-979445

ABSTRACT

ObjectiveTo investigate the effect of Zuoguiwan on pancreatic islet function in offspring of gestational diabetes mellitus (GDM) maternal rat model and explore the mechanisms of Zuoguiwan in improving pancreatic islet function based on postpartum pancreatic regeneration. MethodHealthy female SD rats with normal blood glucose levels were paired with male rats in a 2∶1 ratio and housed together. Pregnancy was confirmed based on vaginal plugs or vaginal smears. The pregnant rats were divided into the following groups: normal group, model group, insulin group (insulin Detemir, 20 U·kg-1), low-dose Zuoguiwan group (1.89 g·kg-1), and high-dose Zuoguiwan group (3.78 g·kg-1). The GDM rat model was induced using streptozotocin in rats except for those in the normal group. The model was confirmed by blood glucose testing in the maternal rats. Except for the normal and model groups, the other groups received daily administration of corresponding treatments. At 21 days after birth, fasting blood glucose (FBG) and fasting serum insulin (FINS) levels were measured in 6 offspring from each group. The homeostasis model assessment of insulin resistance (HOMA-IR) was calculated, and an oral glucose tolerance test (OGTT) was performed on additional 12 offspring from each group. Blood samples were taken from the abdominal aorta of the offspring at postnatal day 22, and enzyme-linked immunosorbent assay (ELISA) was used to measure insulin, glucagon (GC), pancreatic polypeptide (PPY), and somatostatin (SS) levels in the serum. Hematoxylin-eosin (HE) staining was performed to observe pathological changes in the pancreatic tissue of the offspring. Immunofluorescence (IF) was used to observe the area and structure of the pancreatic islets. Western blot was used to detect the expression of key proteins involved in the development and functional expression of pancreatic β-cells, namely pancreatic and duodenal homeobox factor 1 (Pdx1), Nkx6.1, and Glucose transporter 2 (Glut2). ResultCompared with the normal group, the model group showed significant increases in FBG and FINS levels, and HOMA-IR (P<0.01). Compared with the model group, the insulin group showed significant decreases in FBG levels and HOMA-IR (P<0.01), the low-dose Zuoguiwan group showed a significant decrease in FBG levels (P<0.05), and the high-dose Zuoguiwan group showed significant decreases in FBG and FINS levels, and HOMA-IR (P<0.01). Compared with the normal group, the model group showed significant increases in OGTT 60-min blood glucose levels and AUC index (P<0.05, P<0.01). Compared with the model group, the high-dose Zuoguiwan group showed significant decreases in OGTT60-min blood glucose levels and area under the curve(AUC) index (P<0.05, P<0.01). HE staining of pancreatic tissue showed that compared with the normal group, the model group had a reduced number of islets and a loose arrangement of acinar cells. Compared with the model group, the groups with drug treatment showed increased number of islets and a compact arrangement of acinar cells. Compared with the normal group, the model group had significantly increased levels of insulin, GC, PPY, and SS in the serum (P<0.01). Compared with the model group, the low-dose and high-dose Zuoguiwan groups and the insulin group showed significantly decreased serum levels of insulin, GC, PPY, and SS (P<0.05, P<0.01). IF results showed that compared with the normal group, the model group had a significantly lower positive rate of insulin (P<0.05). Compared with the model group, the low-dose and high-dose Zuoguiwan groups showed a significant increase in the positive rate of insulin (P<0.05). There was no significant difference in the positive rate of GC among the groups. In terms of the proportion of insulin and GC in individual islets, compared with the normal group, the model group showed a significant decrease in the proportion of insulin (P<0.01) and a significant increase in the proportion of GC (P<0.01). Compared with the model group, the low-dose and high-dose Zuoguiwan groups showed significantly increased proportion of insulin (P<0.01) and significantly decreased proportion of GC (P<0.01). Compared with the normal group, the model group showed significantly decreased expression levels of Pdx1, Nkx6.1, and Glut2 proteins in the pancreatic tissue of GDM offspring (P<0.05). Compared with the model group, the insulin group and the low-dose Zuoguiwan group showed significant increases in the expression levels of Pdx1 and Nkx6.1 proteins in the pancreatic tissue of GDM offspring (P<0.05), and the low-dose and high-dose Zuoguiwan groups showed significant increases in the expression levels of Glut2 protein (P<0.05). ConclusionZuoguiwan can promote pancreatic islet development in offspring of GDM maternal rat model, improve pancreatic islet morphology and function, and alleviate insulin resistance. Its mechanism of action may be related to the regulation of Pdx1, Nkx6.1, and Glut2 protein expression in the pancreatic tissue of offspring.

11.
Trends Dev Biol ; 16: 1-27, 2023.
Article in English | MEDLINE | ID: mdl-38873037

ABSTRACT

Epigenetic modifications to DNA and its associated proteins affect cell plasticity and cell fate restrictions throughout embryonic development. Development of the vertebrate pancreas is characterized by initial is an over-lapping expression of a set of transcriptional regulators in a defined region of the posterior foregut endoderm that collectively promote pancreas progenitor specification and proliferation. As development progresses, these transcription factors segregate into distinct pancreatic lineages, with some being maintained in specific subsets of terminally differentiated pancreas cell types throughout adulthood. Here we describe the progressive stages and cell fate restrictions that occur during pancreas development and the relevant known epigenetic regulatory events that drive the dynamic expression patterns of transcription factors that regulate pancreas development. In addition, we highlight how changes in epigenetic marks can affect susceptibility to pancreas diseases (such as diabetes), adult pancreas cell plasticity, and the ability to derive replacement insulin-producing ß cells for the treatment of diabetes.

12.
Front Endocrinol (Lausanne) ; 13: 1042611, 2022.
Article in English | MEDLINE | ID: mdl-36339450

ABSTRACT

Diabetes is an epidemic with increasing incidence across the world. Most individuals who are afflicted by this disease have type 2 diabetes, but there are many who suffer from type 1, an autoimmune disorder. Both types of diabetes have complex genetic underpinnings that are further complicated by epigenetic and environmental factors. A less prevalent and often under diagnosed subset of diabetes cases are characterized by single genetic mutations and include Maturity Onset Diabetes of the Young (MODY) and Neonatal Diabetes Mellitus (NDM). While the mode of action and courses of treatment for all forms of diabetes are distinct, the diseases all eventually result in the dysfunction and/or death of the pancreatic ß cell - the body's source of insulin. With loss of ß cell function, blood glucose homeostasis is disrupted, and life-threatening complications arise. In this review, we focus on how model systems provide substantial insights into understanding ß cell biology to inform our understanding of all forms of diabetes. The strengths and weaknesses of animal, hPSC derived ß-like cell, and organoid models are considered along with discussion of GATA6, a critical transcription factor frequently implicated in pancreatic dysfunction with developmental origins; experimental studies of GATA6 have highlighted the advantages and disadvantages of how each of these model systems can be used to inform our understanding of ß cell specification and function in health and disease.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Humans , Mice , Animals , Diabetes Mellitus, Type 2/complications , Organoids , Pancreas , Stem Cells
13.
Animals (Basel) ; 12(19)2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36230305

ABSTRACT

This experiment was conducted to investigate the effect of supplemental concentrate feeding on the pancreatic development of yak calves. Twenty one-month-old yak calves with healthy body condition and similar body weight were selected as experimental animals and randomly divided into two groups, five replicates in each group. The control group yak calves were fed milk replacer and alfalfa hay, the experimental group yak calves were fed milk replacer, alfalfa hay and concentrate. The pre-feeding period of this experiment was thirty days, the trial period was one hundred days. At the end of feeding trail, five yak calves from each group were selected and slaughtered and the pancreas tissues of yak calves were collected and determined. The results showed that: (1) Dry matter and body weight of yak calves in the test group were significantly higher than those of the control group. (2) The apparent nutrient digestibility of crude protein, crude fat, calcium and phosphorus in the test group of yak calves was significantly higher than that of the control group, while the apparent nutrient digestibility of neutral detergent fiber and acid detergent fiber in the test group was significantly lower than that of the control group. (3) Pancreatic weight, organ index, total ratio of exocrine part area and total ratio of endocrine area of yak calves in the test group were significantly higher than those in the control group, while the ratio of exocrine area was significantly lower in the test group than that of the control group. (4) The activities of the main pancreatic digestive enzymes: pancreatic amylase, pancreatic lipase, pancreatic protease and chymotrypsin were significantly higher in the test group than those of the control group, as were the hormonal contents of glucagon, insulin and pancreatic polypeptide. (5) The main differential metabolites of the pancreas in the test group were significantly higher than those of the control group, such as D-proline, hypoxanthine, acetylcysteine, gamma-glutamylcysteine, thiazolidine-4-carboxylic acid, piperidinic acid, ellagic acid, nicotinamide, tropolone, D-serine, ribulose-5-phosphate, (+/-)5(6)-epoxyeicosatrienoic acid(EET), 2-hydroxycinnamic acid, L-phenylalanine, creatinine, tetrahydrocorticosterone, pyridoxamine, xanthine, 5-oxoproline, asparagine, DL-tryptophan, in-dole-3-acrylic acid, thymine, trehalose, docosapentaenoic acid, docosahexaenoic acid, fatty acid esters of hydroxy fatty acids(FAHFA) (18:1/20:3), fatty acid esters of hydroxy fatty acids(FAHFA) (18:2/20:4), adrenic acid and xanthosine. In conclusion, supplemental concentrate feeding promoted the good development of morphological and functional properties of the pancreas in early weaned yak calves to improve the digestion and absorption of feed nutrients, so as to enhance the growth and development quality of early weaned yak calves.

14.
Genes (Basel) ; 13(9)2022 09 10.
Article in English | MEDLINE | ID: mdl-36140793

ABSTRACT

Studies in animal diabetic models have demonstrated the possibility of islet regeneration through treatment with natural extracts, such as Allium sativum (garlic). This study aimed to investigate the effect of garlic extract (GE) on the expression of three genes (Ngn3, Pdx1, and MafA) in the pancreas and liver of diabetic rats. Thirty-two rats were divided into two groups, streptozotocin (STZ)-induced diabetic rats (n = 16) and healthy rats (n = 16). Both groups were subdivided into GE-treated (n = 8), and those administered 0.9% normal saline (NS) (n = 8) for 1 week (n = 4) and 8 weeks (n = 4). In the pancreas of diabetic rats treated with GE for 1 week, all three genes, Ngn3, Pdx1, and MafA, were significantly upregulated (p ≤ 0.01, p ≤ 0.05, and p ≤ 0.001, respectively) when compared to diabetic rats treated with NS only. However, after eight weeks of GE treatment, the expression of all three genes decreased as blood insulin increased. In the liver, only Pdx1 expression significantly (p ≤ 0.05) increased after 8 weeks. The significant expression of Ngn3, Pdx1, and MafA in the pancreas by week 1 may have induced the maturation of juvenile ß-cells, which escaped the effects of STZ and caused an increase in serum insulin.


Subject(s)
Diabetes Mellitus, Experimental , Insulins , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Insulins/metabolism , Liver , Pancreas/metabolism , Rats , Saline Solution/metabolism , Streptozocin
15.
Dev Cell ; 57(16): 1922-1936.e9, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35998583

ABSTRACT

Sequence variants in cis-acting enhancers are important for polygenic disease, but their role in Mendelian disease is poorly understood. Redundancy between enhancers that regulate the same gene is thought to mitigate the pathogenic impact of enhancer mutations. Recent findings, however, have shown that loss-of-function mutations in a single enhancer near PTF1A cause pancreas agenesis and neonatal diabetes. Using mouse and human genetic models, we show that this enhancer activates an entire PTF1A enhancer cluster in early pancreatic multipotent progenitors. This leading role, therefore, precludes functional redundancy. We further demonstrate that transient expression of PTF1A in multipotent progenitors sets in motion an epigenetic cascade that is required for duct and endocrine differentiation. These findings shed insights into the genome regulatory mechanisms that drive pancreas differentiation. Furthermore, they reveal an enhancer that acts as a regulatory master key and is thus vulnerable to pathogenic loss-of-function mutations.


Subject(s)
Diabetes Mellitus , Transcription Factors , Animals , Cell Differentiation/genetics , Diabetes Mellitus/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Mice , Mutation/genetics , Pancreas/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
16.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35708349

ABSTRACT

Pancreatic and duodenal homeobox 1 (PDX1) is crucial for pancreas organogenesis, yet the dynamic changes in PDX1 binding in human or mouse developing pancreas have not been examined. To address this knowledge gap, we performed PDX1 ChIP-seq and single-cell RNA-seq using fetal human pancreata. We integrated our datasets with published datasets and revealed the dynamics of PDX1 binding and potential cell lineage-specific PDX1-bound genes in the pancreas from fetal to adult stages. We identified a core set of developmentally conserved PDX1-bound genes that reveal the broad multifaceted role of PDX1 in pancreas development. Despite the well-known dramatic changes in PDX1 function and expression, we found that PDX1-bound genes are largely conserved from embryonic to adult stages. This points towards a dual role of PDX1 in regulating the expression of its targets at different ages, dependent on other functionally congruent or directly interacting partners. We also showed that PDX1 binding is largely conserved in mouse pancreas. Together, our study reveals PDX1 targets in the developing pancreas in vivo and provides an essential resource for future studies on pancreas development.


Subject(s)
Genes, Homeobox , Homeodomain Proteins , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Pancreas , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome/genetics
17.
Front Endocrinol (Lausanne) ; 13: 904004, 2022.
Article in English | MEDLINE | ID: mdl-35769082

ABSTRACT

Diabetes mellitus, a disease that affects nearly 536.6 million people worldwide, is characterized by the death or dysfunction of insulin-producing beta cells of the pancreas. The beta cells are found within the islets of Langerhans, which are composed of multiple hormone-producing endocrine cells including the alpha (glucagon), delta (somatostatin), PP (pancreatic polypeptide), and epsilon (ghrelin) cells. There is direct evidence that physical and paracrine interactions between the cells in the islet facilitate and support beta cell function. However, communication between endocrine and exocrine cells in the pancreas may also directly impact beta cell growth and function. Herein we review literature that contributes to the view that "crosstalk" between neighboring cells within the pancreas influences beta cell growth and function and the maintenance of beta cell health.


Subject(s)
Diabetes Mellitus , Endocrine Cells , Insulin-Secreting Cells , Glucagon , Humans , Pancreas
18.
Curr Top Dev Biol ; 149: 341-372, 2022.
Article in English | MEDLINE | ID: mdl-35606060

ABSTRACT

Tight spatiotemporal control of cellular behavior and cell fate decisions is paramount to the formation of multicellular organisms during embryonic development. Intercellular communication via signaling pathways mediates this control. Interestingly, these signaling pathways are not static, but dynamic and change in activity over time. Signaling oscillations as a specific type of dynamics are found in various signaling pathways and model systems. Functions of oscillations include the regulation of periodic events or the transmission of information by encoding signals in the dynamic properties of a signaling pathway. For instance, signaling oscillations in neural or pancreatic progenitor cells modulate their proliferation and differentiation. Oscillations between neighboring cells can also be synchronized, leading to the emergence of waves traveling through the tissue. Such population-wide signaling oscillations regulate for example the consecutive segmentation of vertebrate embryos, a process called somitogenesis. Here, we outline our current understanding of signaling oscillations in embryonic development, how signaling oscillations are generated, how they are studied and how they contribute to the regulation of embryonic development.


Subject(s)
Receptors, Notch , Somites , Cell Communication , Embryonic Development/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology
19.
Eur J Pharm Sci ; 172: 106148, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35189271

ABSTRACT

Type 1 diabetes mellitus affects 45 million people worldwide and its prevalence is rapidly increasing. It derives from a lack of insulin production by the pancreas, which leads to elevated blood sugar levels. Current treatments rely on the administration of exogenous insulin, but they do not replicate the precise control of glycemia by the pancreas. Whole pancreas and pancreatic islet transplantation restore endogenous insulin secretion in response to blood glucose levels. However, both are limited by the lack of donors and the need for immunosuppressive therapy. Pluripotent stem cells are a virtually unlimited cell source and can be differentiated to the desired cell types. Moreover, induced pluripotent stem cells may be derived from the patient's cells, which could prevent graft rejection. Several protocols report the differentiation of pluripotent stem cells into insulin-producing cells that, after transplantation, can restore glycemic control. Such protocols are based on the embryonic development of the pancreas, highlighting the importance of understanding the different stages and signaling pathways involved in this process. Once the main hurdles to stem cell-based therapies are overcome, translation to clinical practice will greatly improve the quality of life of people with type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans Transplantation , Pluripotent Stem Cells , Cell Differentiation , Diabetes Mellitus, Type 1/therapy , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Pluripotent Stem Cells/metabolism , Quality of Life
20.
FEBS J ; 289(17): 5121-5136, 2022 09.
Article in English | MEDLINE | ID: mdl-34125483

ABSTRACT

Vertebrate pancreas organogenesis is a stepwise process regulated by a complex network of signaling and transcriptional events, progressively steering the early endoderm toward pancreatic fate. Many crucial players of this process have been identified, including signaling pathways, cis-regulatory elements, and transcription factors (TFs). Pancreas-associated transcription factor 1a (PTF1A) is one such TF, crucial for pancreas development. PTF1A mutations result in dramatic pancreatic phenotypes associated with severe complications, such as neonatal diabetes and impaired food digestion due to exocrine pancreatic insufficiency. Here, we present a brief overview of vertebrate pancreas development, centered on Ptf1a function and transcriptional regulation, covering similarities and divergences in three broadly studied organisms: human, mouse and zebrafish.


Subject(s)
Gene Expression Regulation, Developmental , Zebrafish , Animals , Humans , Mice , Organogenesis/genetics , Pancreas/metabolism , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...