Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 50
1.
Diabetes ; 72(8): 1112-1126, 2023 08 01.
Article En | MEDLINE | ID: mdl-37216637

The loss of pancreatic ß-cell identity has emerged as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell-cycle regulator and transcription factor E2F1 in the maintenance of ß-cell identity, insulin secretion, and glucose homeostasis. We show that the ß-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, altered endocrine cell mass, downregulation of many ß-cell genes, and concomitant increase of non-ß-cell markers. Mechanistically, epigenomic profiling of the promoters of these non-ß-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic, and epigenomic signatures are associated with these ß-cell dysfunctions, with E2F1 directly regulating several ß-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of ß-cell identity genes. Our data suggest that E2F1 is critical for maintaining ß-cell identity and function through sustained control of ß-cell and non-ß-cell transcriptional programs. ARTICLE HIGHLIGHTS: ß-Cell-specific E2f1 deficiency in mice impairs glucose tolerance. Loss of E2f1 function alters the ratio of α- to ß-cells but does not trigger ß-cell conversion into α-cells. Pharmacological inhibition of E2F activity inhibits glucose-stimulated insulin secretion and alters ß- and α-cell gene expression in human islets. E2F1 maintains ß-cell function and identity through control of transcriptomic and epigenetic programs.


Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Humans , Mice , Chromatin/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Histones/metabolism , Homeostasis/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice, Knockout
2.
Cells ; 12(4)2023 02 09.
Article En | MEDLINE | ID: mdl-36831232

The pancreas is an abdominal gland that serves 2 vital purposes: assist food processing by secreting digestive enzymes and regulate blood glucose levels by releasing endocrine hormones. During embryonic development, this gland originates from epithelial buds located on opposite sites of the foregut endoderm. Pancreatic cell specification and maturation are coordinated by a complex interplay of extrinsic and intrinsic signaling events. In the recent years, the canonical Wnt/ß-catenin pathway has emerged as an important player of pancreas organogenesis, regulating pancreatic epithelium specification, compartmentalization and expansion. Importantly, it has been suggested to regulate proliferation, survival and function of adult pancreatic cells, including insulin-secreting ß-cells. This review summarizes recent work on the role of Wnt/ß-catenin signaling in pancreas biology from early development to adulthood, emphasizing on its relevance for the development of new therapies for pancreatic diseases.


Wnt Signaling Pathway , beta Catenin , Pregnancy , Female , Humans , beta Catenin/metabolism , Pancreas/metabolism , Organogenesis , Embryonic Development
3.
Front Endocrinol (Lausanne) ; 13: 861922, 2022.
Article En | MEDLINE | ID: mdl-35573999

Type 1 diabetes results from the autoimmune-mediated loss of insulin-producing beta-cells. Accordingly, important research efforts aim at regenerating these lost beta-cells by converting pre-existing endogenous cells. Following up on previous results demonstrating the conversion of pancreatic somatostatin delta-cells into beta-like cells upon Pax4 misexpression and acknowledging that somatostatin-expressing cells are highly represented in the gastrointestinal tract, one could wonder whether this Pax4-mediated conversion could also occur in the GI tract. We made use of transgenic mice misexpressing Pax4 in somatostatin cells (SSTCrePOE) to evaluate a putative Pax4-mediated D-to-beta-like cell conversion. Additionally, we implemented an ex vivo approach based on mice-derived gut organoids to assess the functionality of these neo-generated beta-like cells. Our results outlined the presence of insulin+ cells expressing several beta-cell markers in gastrointestinal tissues of SSTCrePOE animals. Further, using lineage tracing, we established that these cells arose from D cells. Lastly, functional tests on mice-derived gut organoids established the ability of neo-generated beta-like cells to release insulin upon stimulation. From this study, we conclude that the misexpression of Pax4 in D cells appears sufficient to convert these into functional beta-like cells, thus opening new research avenues in the context of diabetes research.


Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Somatostatin-Secreting Cells , Animals , Homeodomain Proteins/genetics , Insulin , Mice , Paired Box Transcription Factors/genetics , Somatostatin/genetics
4.
Cells ; 10(11)2021 10 20.
Article En | MEDLINE | ID: mdl-34831029

Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on ß-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of ß-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.


DNA-Binding Proteins/deficiency , Diabetes Mellitus/metabolism , Diabetes Mellitus/prevention & control , Transcription Factors/deficiency , Acinar Cells/cytology , Acinar Cells/metabolism , Amylases/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus/genetics , Disease Models, Animal , Gene Expression Regulation , Ghrelin/metabolism , Homeodomain Proteins/metabolism , Hyperglycemia/complications , Hyperglycemia/genetics , Integrases/metabolism , Mice, Transgenic , Mutation/genetics , Pancreas/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
NPJ Regen Med ; 6(1): 7, 2021 Feb 12.
Article En | MEDLINE | ID: mdl-33580013

The role of DNA methylation in ß-cell neogenesis is poorly understood. We report that during the process of induced cell reprogramming, methylation content of the Ngn3 and Sox11 genes are diminished. These findings emphasise DNA methylation is a barrier in ß-cell regeneration in adulthood, a well described pathophysiological phenomenon of major significance in explaining ß-cell deficiency in diabetes in the adult pancreas.

6.
Genome Biol ; 21(1): 106, 2020 05 06.
Article En | MEDLINE | ID: mdl-32375897

BACKGROUND: Single-cell RNA-seq (scRNA-seq) is emerging as a powerful tool to dissect cell-specific effects of drug treatment in complex tissues. This application requires high levels of precision, robustness, and quantitative accuracy-beyond those achievable with existing methods for mainly qualitative single-cell analysis. Here, we establish the use of standardized reference cells as spike-in controls for accurate and robust dissection of single-cell drug responses. RESULTS: We find that contamination by cell-free RNA can constitute up to 20% of reads in human primary tissue samples, and we show that the ensuing biases can be removed effectively using a novel bioinformatics algorithm. Applying our method to both human and mouse pancreatic islets treated ex vivo, we obtain an accurate and quantitative assessment of cell-specific drug effects on the transcriptome. We observe that FOXO inhibition induces dedifferentiation of both alpha and beta cells, while artemether treatment upregulates insulin and other beta cell marker genes in a subset of alpha cells. In beta cells, dedifferentiation and insulin repression upon artemether treatment occurs predominantly in mouse but not in human samples. CONCLUSIONS: This new method for quantitative, error-correcting, scRNA-seq data normalization using spike-in reference cells helps clarify complex cell-specific effects of pharmacological perturbations with single-cell resolution and high quantitative accuracy.


Islets of Langerhans/drug effects , RNA-Seq/standards , Single-Cell Analysis/standards , Animals , Artemether/pharmacology , Cell Dedifferentiation/drug effects , Forkhead Transcription Factors/antagonists & inhibitors , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Machine Learning , Mice , Reference Standards , Species Specificity , Transcriptome/drug effects
7.
Hum Mol Genet ; 28(24): 4089-4102, 2019 12 15.
Article En | MEDLINE | ID: mdl-31691806

A disproportional large number of neurodevelopmental disorders (NDDs) is caused by variants in genes encoding transcription factors and chromatin modifiers. However, the functional interactions between the corresponding proteins are only partly known. Here, we show that KDM5C, encoding a H3K4 demethylase, is at the intersection of transcriptional axes under the control of three regulatory proteins ARX, ZNF711 and PHF8. Interestingly, mutations in all four genes (KDM5C, ARX, ZNF711 and PHF8) are associated with X-linked NDDs comprising intellectual disability as a core feature. in vitro analysis of the KDM5C promoter revealed that ARX and ZNF711 function as antagonist transcription factors that activate KDM5C expression and compete for the recruitment of PHF8. Functional analysis of mutations in these genes showed a correlation between phenotype severity and the reduction in KDM5C transcriptional activity. The KDM5C decrease was associated with a lack of repression of downstream target genes Scn2a, Syn1 and Bdnf in the embryonic brain of Arx-null mice. Aiming to correct the faulty expression of KDM5C, we studied the effect of the FDA-approved histone deacetylase inhibitor suberanilohydroxamic acid (SAHA). In Arx-KO murine ES-derived neurons, SAHA was able to rescue KDM5C depletion, recover H3K4me3 signalling and improve neuronal differentiation. Indeed, in ARX/alr-1-deficient Caenorhabditis elegans animals, SAHA was shown to counteract the defective KDM5C/rbr-2-H3K4me3 signalling, recover abnormal behavioural phenotype and ameliorate neuronal maturation. Overall, our studies indicate that KDM5C is a conserved and druggable effector molecule across a number of NDDs for whom the use of SAHA may be considered a potential therapeutic strategy.


Histone Demethylases/metabolism , Neurodevelopmental Disorders/metabolism , Animals , Caenorhabditis elegans , Cell Line , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurodevelopmental Disorders/genetics , Neurons/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/metabolism , Vorinostat/pharmacology
8.
Diabetes Obes Metab ; 20 Suppl 2: 3-10, 2018 09.
Article En | MEDLINE | ID: mdl-30230184

Ghrelin is a gastric peptide with anabolic functions. It acutely stimulates growth hormone (GH) secretion from the anterior pituitary glands and modulates hypothalamic circuits that control food intake and energy expenditure. Besides its central activity, ghrelin is also involved in the regulation of pancreatic development and physiology. Particularly, several studies highlighted the ability of ghrelin to sustain ß-cell viability and proliferation. Furthermore, ghrelin seems to exert inhibitory effects on pancreatic acinar and endocrine secretory functions. Due to its pleiotropic activity on energy metabolism, ghrelin has become a topic of great interest for experimental research focused on type II diabetes and obesity. The aim of this review is to illustrate the complex and not fully understood interplay between ghrelin, pancreas and glucose homeostasis.


Ghrelin/physiology , Pancreas/growth & development , Animals , Blood Glucose/metabolism , Diabetes Mellitus/etiology , Ghrelin/genetics , Homeostasis/physiology , Humans , Hypothalamus/physiology , Mice , Pancreas/physiology , Receptors, Ghrelin/physiology
9.
Genes (Basel) ; 9(9)2018 Sep 07.
Article En | MEDLINE | ID: mdl-30205460

Both type 1 and type 2 diabetes are conditions that are associated with the loss of insulin-producing ß-cells within the pancreas. An active research therefore aims at regenerating these ß-cells with the hope that they could restore euglycemia. The approaches classically used consist in mimicking embryonic development, making use of diverse cell sources or converting pre-existing pancreatic cells. Despite impressive progresses and promising successes, it appears that we still need to gain further insight into the molecular mechanisms underlying ß-cell development. This becomes even more obvious with the emergence of a relatively new field of research, epigenetics. The current review therefore focuses on the latest advances in this field in the context of ß-cell (neo-)genesis research.

10.
PLoS One ; 13(8): e0201536, 2018.
Article En | MEDLINE | ID: mdl-30092080

In the context of type 1 diabetes research and the development of insulin-producing ß-cell replacement strategies, whether pancreatic ductal cells retain their developmental capability to adopt an endocrine cell identity remains debated, most likely due to the diversity of models employed to induce pancreatic regeneration. In this work, rather than injuring the pancreas, we developed a mouse model allowing the inducible misexpression of the proendocrine gene Neurog3 in ductal cells in vivo. These animals developed a progressive islet hypertrophy attributed to a proportional increase in all endocrine cell populations. Lineage tracing experiments indicated a continuous neo-generation of endocrine cells exhibiting a ductal ontogeny. Interestingly, the resulting supplementary ß-like cells were found to be functional. Based on these findings, we suggest that ductal cells could represent a renewable source of new ß-like cells and that strategies aiming at controlling the expression of Neurog3, or of its molecular targets/co-factors, may pave new avenues for the improved treatments of diabetes.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Plasticity/physiology , Diabetes Mellitus, Type 1/pathology , Endocrine Cells/physiology , Nerve Tissue Proteins/metabolism , Pancreatic Ducts/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Pancreatic Ducts/cytology , Regeneration
12.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Article En | MEDLINE | ID: mdl-29025873

The recent demonstration that pancreatic α cells can be continuously regenerated and converted into ß-like cells upon ectopic expression of Pax4 opened new avenues of research in the endocrine cell differentiation and diabetes fields. To determine whether such plasticity was also shared by δ cells, we generated and characterized transgenic animals that express Pax4 specifically in somatostatin-expressing cells. We demonstrate that the ectopic expression of Pax4 in δ cells is sufficient to induce their conversion into functional ß-like cells. Importantly, this conversion induces compensatory mechanisms involving the reactivation of endocrine developmental processes that result in dramatic ß-like cell hyperplasia. Importantly, these ß-like cells are functional and can partly reverse the consequences of chemically induced diabetes.


Diabetes Mellitus, Experimental/genetics , Ectopic Gene Expression , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Cell Proliferation , Cell Transdifferentiation/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Genetic Therapy/methods , Glucagon/biosynthesis , Glucagon/genetics , Homeodomain Proteins/metabolism , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Transgenic , Paired Box Transcription Factors/metabolism , Somatostatin/biosynthesis , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Streptozocin
13.
Commun Integr Biol ; 10(3): e1300215, 2017.
Article En | MEDLINE | ID: mdl-28702122

Diabetes is a chronic and progressing disease, the number of patients increasing exponentially, especially in industrialized countries. Regenerating lost insulin-producing cells would represent a promising therapeutic alternative for most diabetic patients. To this end, using the mouse as a model, we reported that GABA, a food supplement, could induce insulin-producing beta-like cell neogenesis offering an attractive and innovative approach for diabetes therapeutics.

14.
Front Genet ; 8: 75, 2017.
Article En | MEDLINE | ID: mdl-28634486

Type 1 diabetes is an auto-immune disease resulting in the loss of pancreatic ß-cells and, consequently, in chronic hyperglycemia. Insulin supplementation allows diabetic patients to control their glycaemia quite efficiently, but treated patients still display an overall shortened life expectancy and an altered quality of life as compared to their healthy counterparts. In this context and due to the ever increasing number of diabetics, establishing alternative therapies has become a crucial research goal. Most current efforts therefore aim at generating fully functional insulin-secreting ß-like cells using multiple approaches. In this review, we screened the literature published since 2011 and inventoried the selected markers used to characterize insulin-secreting cells generated by in vitro differentiation of stem/precursor cells or by means of in vivo transdifferentiation. By listing these features, we noted important discrepancies when comparing the different approaches for the initial characterization of insulin-producing cells as true ß-cells. Considering the recent advances achieved in this field of research, the necessity to establish strict guidelines has become a subject of crucial importance, especially should one contemplate the next step, which is the transplantation of in vitro or ex vivo generated insulin-secreting cells in type 1 diabetic patients.

16.
Cell ; 168(1-2): 73-85.e11, 2017 Jan 12.
Article En | MEDLINE | ID: mdl-27916274

The recent discovery that genetically modified α cells can regenerate and convert into ß-like cells in vivo holds great promise for diabetes research. However, to eventually translate these findings to human, it is crucial to discover compounds with similar activities. Herein, we report the identification of GABA as an inducer of α-to-ß-like cell conversion in vivo. This conversion induces α cell replacement mechanisms through the mobilization of duct-lining precursor cells that adopt an α cell identity prior to being converted into ß-like cells, solely upon sustained GABA exposure. Importantly, these neo-generated ß-like cells are functional and can repeatedly reverse chemically induced diabetes in vivo. Similarly, the treatment of transplanted human islets with GABA results in a loss of α cells and a concomitant increase in ß-like cell counts, suggestive of α-to-ß-like cell conversion processes also in humans. This newly discovered GABA-induced α cell-mediated ß-like cell neogenesis could therefore represent an unprecedented hope toward improved therapies for diabetes.


Diabetes Mellitus/drug therapy , Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , gamma-Aminobutyric Acid/administration & dosage , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucagon-Secreting Cells/drug effects , Humans , Islets of Langerhans/cytology , Male , Mice , Nerve Tissue Proteins , Rats , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology
17.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article En | MEDLINE | ID: mdl-27916275

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
18.
PLoS One ; 10(12): e0144597, 2015.
Article En | MEDLINE | ID: mdl-26658466

The transcription factor Pax6 is an important regulator of development and cell differentiation in various organs. Thus, Pax6 was shown to promote neural development in the cerebral cortex and spinal cord, and to control pancreatic endocrine cell genesis. However, the role of Pax6 in distinct endocrine cells of the adult pancreas has not been addressed. We report the conditional inactivation of Pax6 in insulin and glucagon producing cells of the adult mouse pancreas. In the absence of Pax6, beta- and alpha-cells lose their molecular maturation characteristics. Our findings provide strong evidence that Pax6 is responsible for the maturation of beta-, and alpha-cells, but not of delta-, and PP-cells. Moreover, lineage-tracing experiments demonstrate that Pax6-deficient beta- and alpha-cells are shunted towards ghrelin marked cells, sustaining the idea that ghrelin may represent a marker for endocrine cell maturation.


Eye Proteins/genetics , Ghrelin/genetics , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Pancreatic Polypeptide-Secreting Cells/metabolism , Repressor Proteins/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Cell Lineage/drug effects , Cell Lineage/genetics , Crosses, Genetic , Eye Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Ghrelin/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Knockout , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Repressor Proteins/metabolism , Signal Transduction , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/drug effects , Tamoxifen/pharmacology
19.
Semin Cell Dev Biol ; 44: 107-14, 2015 Aug.
Article En | MEDLINE | ID: mdl-26319183

The embryonic development of the pancreas is orchestrated by a complex and coordinated transcription factor network. Neurogenin3 (Neurog3) initiates the endocrine program by activating the expression of additional transcription factors driving survival, proliferation, maturation and lineage allocation of endocrine precursors. Among the direct targets of Neurog3, Pax4 appears as one of the key regulators of ß-cell specification. Indeed, mice lacking Pax4 die a few days postpartum, as they develop severe hyperglycemia due to the absence of mature pancreatic ß-cells. Pax4 also directly regulates the expression of Arx, a gene that plays a crucial role in α-cell specification. Comparative analysis of Pax4 and Arx mutants, as well as Arx/Pax4 double mutants, showed that islet subtype destiny is mainly directed by cross-repression of the Pax4 and Arx factors. Importantly, the ectopic expression of Pax4 in α-cells was found sufficient to induce their neogenesis and conversion into ß-like cells, not only during development but also in adult rodents. Therefore, differentiated endocrine α-cells can be considered as a putative source for insulin-producing ß-like cells. These findings have clearly widened our understanding regarding pancreatic development, but they also open new research avenues in the context of diabetes research.


Homeodomain Proteins/physiology , Paired Box Transcription Factors/physiology , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreas/embryology , Pancreas/metabolism , Pancreas/physiology
20.
World J Stem Cells ; 6(2): 248-55, 2014 Apr 26.
Article En | MEDLINE | ID: mdl-24772251

AIM: To find a safe source for dopaminergic neurons, we generated neural progenitor cell lines from human embryonic stem cells. METHODS: The human embryonic stem (hES) cell line H9 was used to generate human neural progenitor (HNP) cell lines. The resulting HNP cell lines were differentiated into dopaminergic neurons and analyzed by quantitative real-time polymerase chain reaction and immunofluorescence for the expression of neuronal differentiation markers, including beta-III tubulin (TUJ1) and tyrosine hydroxylase (TH). To assess the risk of teratoma or other tumor formation, HNP cell lines and mouse neuronal progenitor (MNP) cell lines were injected subcutaneously into immunodeficient SCID/beige mice. RESULTS: We developed a fairly simple and fast protocol to obtain HNP cell lines from hES cells. These cell lines, which can be stored in liquid nitrogen for several years, have the potential to differentiate in vitro into dopaminergic neurons. Following day 30 of differentiation culture, the majority of the cells analyzed expressed the neuronal marker TUJ1 and a high proportion of these cells were positive for TH, indicating differentiation into dopaminergic neurons. In contrast to H9 ES cells, the HNP cell lines did not form tumors in immunodeficient SCID/beige mice within 6 mo after subcutaneous injection. Similarly, no tumors developed after injection of MNP cells. Notably, mouse ES cells or neuronal cells directly differentiated from mouse ES cells formed teratomas in more than 90% of the recipients. CONCLUSION: Our findings indicate that neural progenitor cell lines can differentiate into dopaminergic neurons and bear no risk of generating teratomas or other tumors in immunodeficient mice.

...