Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Front Endocrinol (Lausanne) ; 11: 563267, 2020.
Article in English | MEDLINE | ID: mdl-33101198

ABSTRACT

Epidemiologic analyses have shed light on an association between type 2 diabetes (T2D) and pancreatic ductal adenocarcinoma (PDAC). Recent data also suggest a potential relationship between T2D and insulinoma. Under rare circumstances, type 1 diabetes (T1D) can also be implicated in tumorigenesis. The biological mechanisms underlying such relationships are extremely complex. Some genetic factors contributing to the development of T2D are shared with pancreatic exocrine and endocrine tumors. Obesity and overweight can also contribute to the initiation and severity of T2D, while aging may influence both endocrine and exocrine tumors. Finally, pharmacological treatments of T2D may have an impact on PDAC. On the other hand, some treatments for insulinoma can trigger diabetes. In the present minireview, we discuss the cellular and molecular mechanisms that could explain these interactions. This analysis may help to define new potential therapeutic strategies.


Subject(s)
Aging/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulinoma/metabolism , Obesity/metabolism , Pancreatic Neoplasms/metabolism , Aging/genetics , Aging/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Humans , Insulinoma/genetics , Insulinoma/pathology , Obesity/genetics , Obesity/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Risk Factors
3.
Diabetes ; 67(1): 78-84, 2018 01.
Article in English | MEDLINE | ID: mdl-29079704

ABSTRACT

The mitochondrial carrier uncoupling protein (UCP) 2 belongs to the family of the UCPs. Despite its name, it is now accepted that UCP2 is rather a metabolite transporter than a UCP. UCP2 can regulate oxidative stress and/or energetic metabolism. In rodents, UCP2 is involved in the control of α- and ß-cell mass as well as insulin and glucagon secretion. Our aim was to determine whether the effects of UCP2 observed on ß-cell mass have an embryonic origin. Thus, we used Ucp2 knockout mice. We found an increased size of the pancreas in Ucp2-/- fetuses at embryonic day 16.5, associated with a higher number of α- and ß-cells. This phenotype was caused by an increase of PDX1+ progenitor cells. Perinatally, an increase in the proliferation of endocrine cells also participates in their expansion. Next, we analyzed the oxidative stress in the pancreata. We quantified an increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2) in the mutant, suggesting an increased production of reactive oxygen species (ROS). Phosphorylation of AKT, an ROS target, was also activated in the Ucp2-/- pancreata. Finally, administration of the antioxidant N-acetyl-l-cysteine to Ucp2-/- pregnant mice alleviated the effect of knocking out UCP2 on pancreas development. Together, these data demonstrate that UCP2 controls pancreas development through the ROS-AKT signaling pathway.


Subject(s)
Pancreas/enzymology , Pancreas/metabolism , Uncoupling Protein 2/metabolism , Animals , Blotting, Western , Cells, Cultured , Glucagon-Secreting Cells/metabolism , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Phosphorylation/genetics , Phosphorylation/physiology , Polymerase Chain Reaction , Reactive Oxygen Species/metabolism , Uncoupling Protein 2/genetics
4.
Cell Med ; 8(3): 99-112, 2016 Dec 03.
Article in English | MEDLINE | ID: mdl-28003935

ABSTRACT

Innovative treatments to cure type 1 diabetes are being actively researched. Among the different strategies, the replacement of ß-cells has given promising results. Classically, islets from cadaveric donors are transplanted into diabetic patients, but recently phase I clinical trials that use stem cell-derived ß-cells have been started. Such protocols require either an immunosuppressive treatment or the macroencapsulation of the ß-cells. They involve cell aggregation and the exposure of the cells to hypoxia. Using an engineered human ß-cell, we have addressed these two problems: a novel human ß-cell line called EndoC-ßH3 was cultured as single cells or aggregated clusters. EndoC-ßH3 cells were also cultured at normal atmospheric oxygen tension (pO2 = 21%) or hypoxia (pO2 = 3%) in the presence or absence of modulators of the hypoxia-inducible factor 1α (HIF1α) pathway. Cell aggregation improved glucose-stimulated insulin secretion, demonstrating the benefit of cell-cell contacts. Low oxygen tension decreased ß-cell viability and their sensitivity to glucose, but did not alter insulin production nor the insulin secretion capacity of the remaining cells. To investigate the role of HIF1α, we first used a HIF stabilizer at pO2 = 21%. This led to a mild decrease in cell viability, impaired glucose sensitivity, and altered insulin secretion. Finally, we used a HIF inhibitor on EndoC-ßH3 pseudoislets exposed to hypoxia. Such treatment considerably decreased cell viability. In conclusion, aggregation of the EndoC-ßH3 cells seems to be important to improve their function. A fraction of the EndoC-ßH3 cells are resistant to hypoxia, depending on the level of activity of HIF1α. Thus, these cells represent a good human cell model for future investigations on islet cell transplantation analysis.

5.
Diabetologia ; 57(11): 2348-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25186293

ABSTRACT

AIM/HYPOTHESIS: Different studies have linked hypoxia to embryonic development. Specifically, when embryonic pancreases are cultured ex vivo under hypoxic conditions (3% O2), beta cell development is impaired. Different cellular signalling pathways are involved in adaptation to hypoxia, including the ubiquitous hypoxia-inducible-factor 1-α (HIF1-α) pathway. We aimed to analyse the effects of HIF1-α stabilisation on fetal pancreas development in vivo. METHODS: We deleted the Vhl gene, which encodes von Hippel-Lindau protein (pVHL), a factor necessary for HIF1-α degradation, by crossing Vhl-floxed mice with Sox9-Cre mice. RESULTS: HIF1-α was stabilised in pancreatic progenitor cells in which the HIF pathway was induced. The number of neurogenin-3 (NGN3)-expressing cells was reduced and consequently endocrine development was altered in Vhl knockout pancreases. HIF1-α stabilisation induced Vegfa upregulation, leading to increased vascularisation. To investigate the impact of increased vascularisation on NGN3 expression, we used a bioassay in which Vhl mutant pancreases were cultured with or without vascular endothelial growth factor (VEGF) receptor 2 (VEGF-R2) inhibitors (e.g. Ki8751). Ex vivo analysis showed that Vhl knockout pancreases developed fewer NGN3-positive cells compared with controls. Interestingly, this effect was blocked when vascularisation was inhibited in the presence of VEGF-R2 inhibitors. CONCLUSIONS/INTERPRETATION: Our data demonstrate that HIF1-α negatively controls beta cell differentiation in vivo by regulating NGN3 expression, and that this effect is mediated by signals from blood vessels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreas/cytology , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Female , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/embryology , Pregnancy , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
7.
Med Sci (Paris) ; 29(8-9): 744-8, 2013.
Article in French | MEDLINE | ID: mdl-24005629

ABSTRACT

Stem cells represent an important tool for the medicine of the future. They are recognized by two main characteristics: they can divide to produce two identical daughter cells (self-renewal) and can differentiate in several cell types (multipotency). Considering these possibilities, stem cells constitute a unique material for tissular regeneration. In the case of pancreas, several types of stem cells have been intensively studied: embryonic stem cells (ES), induced pluripotent stem cells (iPS) and adult stem cells. In each case, several strategies have been used to define their identity and to characterize the signals controlling their proliferation and their differentiation into functional insulin-secreting cells. It seems now necessary to determine what the proportion of Myth and Reality is.


Subject(s)
Diabetes Mellitus/surgery , Stem Cell Transplantation/methods , Adult Stem Cells/transplantation , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/transplantation , Humans , Induced Pluripotent Stem Cells/transplantation , Insulin-Secreting Cells/physiology
8.
PLoS Genet ; 9(1): e1003182, 2013.
Article in English | MEDLINE | ID: mdl-23300486

ABSTRACT

Cyanide-resistant non-phosphorylating respiration is known in mitochondria from plants, fungi, and microorganisms but is absent in mammals. It results from the activity of an alternative oxidase (AOX) that conveys electrons directly from the respiratory chain (RC) ubiquinol pool to oxygen. AOX thus provides a bypath that releases constraints on the cytochrome pathway and prevents the over-reduction of the ubiquinone pool, a major source of superoxide. RC dysfunctions and deleterious superoxide overproduction are recurrent themes in human pathologies, ranging from neurodegenerative diseases to cancer, and may be instrumental in ageing. Thus, preventing RC blockade and excess superoxide production by means of AOX should be of considerable interest. However, because of its energy-dissipating properties, AOX might produce deleterious effects of its own in mammals. Here we show that AOX can be safely expressed in the mouse (MitAOX), with major physiological parameters being unaffected. It neither disrupted the activity of other RC components nor decreased oxidative phosphorylation in isolated mitochondria. It conferred cyanide-resistance to mitochondrial substrate oxidation and decreased reactive oxygen species (ROS) production upon RC blockade. Accordingly, AOX expression was able to support cyanide-resistant respiration by intact organs and to afford prolonged protection against a lethal concentration of gaseous cyanide in whole animals. Taken together, these results indicate that AOX expression in the mouse is innocuous and permits to overcome a RC blockade, while reducing associated oxidative insult. Therefore, the MitAOX mice represent a valuable tool in order to investigate the ability of AOX to counteract the panoply of mitochondrial-inherited diseases originating from oxidative phosphorylation defects.


Subject(s)
Electron Transport Complex IV , Mitochondria , Oxidoreductases , Reactive Oxygen Species , Animals , Ciona intestinalis/genetics , Electron Transport/genetics , Electron Transport/physiology , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/physiology , Oxidation-Reduction , Oxidative Phosphorylation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
9.
FASEB J ; 26(7): 2734-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22426121

ABSTRACT

During early embryogenesis, the pancreas shows a paucity of blood flow, and oxygen tension, the partial pressure of oxygen (pO(2)), is low. Later, the blood flow increases as ß-cell differentiation occurs. We have previously reported that pO(2) controls ß-cell development in rats. Here, we checked that hypoxia inducible factor 1α (HIF1α) is essential for this control. First, we demonstrated that the effect of pO(2) on ß-cell differentiation in vitro was independent of epitheliomesenchymal interactions and that neither oxidative nor energetic stress occurred. Second, the effect of pO(2) on pancreas development was shown to be conserved among species, since increasing pO(2) to 21 vs. 3% also induced ß-cell differentiation in mouse (7-fold, P<0.001) and human fetal pancreas. Third, the effect of hypoxia was mediated by HIF1α, since the addition of an HIF1α inhibitor at 3% O(2) increased the number of NGN3-expressing progenitors as compared to nontreated controls (9.2-fold, P<0.001). In contrast, when we stabilized HIF1α by deleting ex vivo the gene encoding pVHL in E13.5 pancreas from Vhl floxed mice, Ngn3 expression and ß-cell development decreased in such Vhl-deleted pancreas compared to controls (2.5 fold, P<0.05, and 6.6-fold, P<0.001, respectively). Taken together, these data demonstrate that HIF1α exerts a negative control over ß-cell differentiation.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA Primers/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Energy Metabolism , Female , Gene Expression Regulation, Developmental , Humans , Hypoxia/embryology , Hypoxia/pathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , In Vitro Techniques , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Islets of Langerhans/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Oxidative Stress , Pregnancy , Rats , Rats, Wistar , Signal Transduction , Species Specificity , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/physiology
10.
PLoS One ; 7(1): e29807, 2012.
Article in English | MEDLINE | ID: mdl-22235342

ABSTRACT

BACKGROUND: Glucose increases the expression of glycolytic enzymes and other hypoxia-response genes in pancreatic beta-cells. Here, we tested whether this effect results from the activation of Hypoxia-Inducible-factors (HIF) 1 and 2 in a hypoxia-dependent manner. METHODOLOGY/PRINCIPAL FINDINGS: Isolated rat islets and insulin-secreting INS-1E cells were stimulated with nutrients at various pO2 values or treated with the HIF activator CoCl2. HIF-target gene mRNA levels and HIF subunit protein levels were measured by real-time RT-PCR, Western Blot and immunohistochemistry. The formation of pimonidazole-protein adducts was used as an indicator of hypoxia. In INS-1E and islet beta-cells, glucose concentration-dependently stimulated formation of pimonidazole-protein adducts, HIF1 and HIF2 nuclear expression and HIF-target gene mRNA levels to a lesser extent than CoCl2 or a four-fold reduction in pO2. Islets also showed signs of HIF activation in diabetic Lepr(db/db) but not non-diabetic Lepr(db/+) mice. In vitro, these glucose effects were reproduced by nutrient secretagogues that bypass glycolysis, and were inhibited by a three-fold increase in pO2 or by inhibitors of Ca²âº influx and insulin secretion. In INS-1E cells, small interfering RNA-mediated knockdown of Hif1α and Hif2α, alone or in combination, indicated that the stimulation of glycolytic enzyme mRNA levels depended on both HIF isoforms while the vasodilating peptide adrenomedullin was a HIF2-specific target gene. CONCLUSIONS/SIGNIFICANCE: Glucose-induced O2 consumption creates an intracellular hypoxia that activates HIF1 and HIF2 in rat beta-cells, and this glucose effect contributes, together with the activation of other transcription factors, to the glucose stimulation of expression of some glycolytic enzymes and other hypoxia response genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucose/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Oxygen/metabolism , Adrenomedullin/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Calcium/metabolism , Cell Hypoxia/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glycolysis/genetics , Hypoxia-Inducible Factor 1/deficiency , Hypoxia-Inducible Factor 1/genetics , Insulin Secretion , Insulin-Secreting Cells/pathology , Kinetics , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
11.
Diabetes ; 61(2): 409-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22210321

ABSTRACT

Leucine (Leu) is an essential branched-chain amino acid, which activates the mammalian target of rapamycin (mTOR) signaling pathway. The effect of Leu on cell differentiation during embryonic development is unknown. Here, we show that Leu supplementation during pregnancy significantly increased fetal body weight, caused fetal hyperglycemia and hypoinsulinemia, and decreased the relative islet area. We also used rat embryonic pancreatic explant culture for elucidating the mechanism of Leu action on ß-cell development. We found that in the presence of Leu, differentiation of pancreatic duodenal homeobox-1-positive progenitor cells into neurogenin3-positive endocrine progenitor cells was inefficient and resulted in decreased ß-cell formation. Mechanistically, Leu increases the intracellular levels of hypoxia-inducible factor 1-α, a repressor of endocrine fate in the pancreas, by activating the mTOR complex 1 signaling pathway. Collectively, our findings indicate that Leu supplementation during pregnancy could potentially increase the risk of type 2 diabetes mellitus by inhibiting the differentiation of pancreatic endocrine progenitor cells during a susceptible period of fetal life.


Subject(s)
Cell Differentiation/drug effects , Insulin-Secreting Cells/drug effects , Leucine/administration & dosage , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Dietary Supplements , Female , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Nerve Tissue Proteins/analysis , Pregnancy , Rats , Rats, Wistar , Sirolimus/pharmacology , Stem Cells/drug effects , Trans-Activators , Transcription Factors/physiology
12.
Diabetes ; 59(3): 662-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20009089

ABSTRACT

OBJECTIVE: Recent evidence indicates that low oxygen tension (pO2) or hypoxia controls the differentiation of several cell types during development. Variations of pO2 are mediated through the hypoxia-inducible factor (HIF), a crucial mediator of the adaptative response of cells to hypoxia. The aim of this study was to investigate the role of pO2 in beta-cell differentiation. RESEARCH DESIGN AND METHODS: We analyzed the capacity of beta-cell differentiation in the rat embryonic pancreas using two in vitro assays. Pancreata were cultured either in collagen or on a filter at the air/liquid interface with various pO2. An inhibitor of the prolyl hydroxylases, dimethyloxaloylglycine (DMOG), was used to stabilize HIF1alpha protein in normoxia. RESULTS: When cultured in collagen, embryonic pancreatic cells were hypoxic and expressed HIF1alpha and rare beta-cells differentiated. In pancreata cultured on filter (normoxia), HIF1alpha expression decreased and numerous beta-cells developed. During pancreas development, HIF1alpha levels were elevated at early stages and decreased with time. To determine the effect of pO2 on beta-cell differentiation, pancreata were cultured in collagen at increasing concentrations of O2. Such conditions repressed HIF1alpha expression, fostered development of Ngn3-positive endocrine progenitors, and induced beta-cell differentiation by O2 in a dose-dependent manner. By contrast, forced expression of HIF1alpha in normoxia using DMOG repressed Ngn3 expression and blocked beta-cell development. Finally, hypoxia requires hairy and enhancer of split (HES)1 expression to repress beta-cell differentiation. CONCLUSIONS: These data demonstrate that beta-cell differentiation is controlled by pO2 through HIF1alpha. Modifying pO2 should now be tested in protocols aiming to differentiate beta-cells from embryonic stem cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Oxygen/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Oxygen/pharmacology , Pregnancy , Rats , Rats, Wistar , Signal Transduction/physiology , Transcription Factor HES-1
13.
J Endocrinol ; 199(2): 287-98, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18772165

ABSTRACT

Mutations of the multiple endocrine neoplasia type 1 (MEN1) gene predispose patients to MEN1 that affects mainly endocrine tissues, suggesting important physiological functions of the gene in adult endocrine cells. Homozygous disruption of Men1 in mice causes embryonic lethality, whereas the eventual involvement of the gene in embryonic development of the endocrine cells remains unknown. Here, we show that homozygous Men1 knockout mice demonstrate a reduced number of glucagon-positive cells in the E12.5 pancreatic bud associated with apoptosis, whereas the exocrine pancreas development in these mice is not affected. Our data suggest that menin is involved in the survival of the early pancreatic endocrine cells during the first developmental transition. Furthermore, chimerism assay revealed that menin has an autonomous and specific effect on the development of islet cells. In addition, using pancreatic bud culture mimicking the differentiation of alpha- and beta-cells during the second transition, we show that loss of menin leads to the failure of endocrine cell development, altered pancreatic structure and a markedly decreased number of cells expressing neurogenin 3, indicating that menin is also required at this stage of the endocrine pancreas development. Taken together, our results suggest that menin plays an indispensable role in the development of the pancreatic endocrine cells.


Subject(s)
Endocrine Cells/cytology , Endocrine Cells/metabolism , Pancreas/metabolism , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , In Vitro Techniques , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/embryology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Biochem Soc Trans ; 36(Pt 3): 276-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18481941

ABSTRACT

Understanding pancreatic development is important for at least three reasons: first, from a cognitive point of view, to understand the development of a complex organ, the pancreas; next, because it is now clear that abnormal pancreatic development can give rise to specific forms of diabetes in humans; and finally, because, if we want to define new treatments for diabetes based on cell therapy or regenerative medicine, we will have to understand in detail how beta-cells develop. In the present paper, we summarize what we currently know concerning pancreatic development and concentrate on some intercellular and environmental signals controlling pancreatic development.


Subject(s)
Pancreas/embryology , Signal Transduction , Animals , Environment , Humans , Transcription Factors/metabolism
15.
Endocr Dev ; 12: 46-54, 2007.
Article in English | MEDLINE | ID: mdl-17923768

ABSTRACT

Over the last decades, pancreas development has been widely investigated. Understanding the mechanisms that control beta-cell development should allow progress towards the regeneration of these cells in humans. Particularly, it is well established that inductive signals from the mesenchyme play an essential role in the proliferation of precursor cells. In the present review, we focused on the roles of fibroblast growth factors (FGFs) in pancreas development. Improvements of the in vivo and in vitro techniques were used to define the function of FGF10. Experiments on FGF10 knockout mice showed that FGF10 is required for the proliferation of precursor cells and the pancreas development. Several laboratories used different in vitro techniques to study the effect of FGF10 on beta-cell differentiation. These methods of investigation are described here. In our experiments, pancreases were placed at the air-liquid interface to define the precise mechanism of action of FGF10. We showed that FGF10 positively regulates the beta-cell mass by increasing the proliferation of the early precursors and by extending the window of expression of the endocrine precursor marker Ngn3. These data are compared with studies performed with other culture systems. Finally, the role of other FGFs is discussed.


Subject(s)
Islets of Langerhans/physiology , Pancreas/embryology , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Fibroblast Growth Factor 10/deficiency , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/physiology , Immunohistochemistry , In Situ Hybridization , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Organ Culture Techniques , Rats , Rats, Mutant Strains
16.
Diabetes ; 56(5): 1248-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17322477

ABSTRACT

The importance of mesenchymal-epithelial interactions for normal development of the pancreas was recognized in the early 1960s, and mesenchymal signals have been shown to control the proliferation of early pancreatic progenitor cells. The mechanisms by which the mesenchyme coordinates cell proliferation and differentiation to produce the normal number of differentiated pancreatic cells are not fully understood. Here, we demonstrate that the mesenchyme positively controls the final number of beta-cells that develop from early pancreatic progenitor cells. In vitro, the number of beta-cells that developed from rat embryonic pancreatic epithelia was larger in cultures with mesenchyme than without mesenchyme. The effect of mesenchyme was not due to an increase in beta-cell proliferation but was due to increased proliferation of early pancreatic duodenal homeobox-1 (PDX1)-positive progenitor cells, as confirmed by bromodeoxyuridine incorporation. Consequently, the window during which early PDX1(+) pancreatic progenitor cells differentiated into endocrine progenitor cells expressing Ngn3 was extended. Fibroblast growth factor 10 mimicked mesenchyme effects on proliferation of early PDX1(+) progenitor cells and induction of Ngn3 expression. Taken together, our results indicate that expansion of early PDX1(+) pancreatic progenitor cells represents a way to increase the final number of beta-cells developing from early embryonic pancreas.


Subject(s)
Epithelial Cells/physiology , Insulin-Secreting Cells/physiology , Pancreas/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Embryo, Mammalian , Epithelial Cells/cytology , Fibroblast Growth Factor 10/pharmacology , Homeostasis , In Situ Hybridization , Insulin/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Pancreas/enzymology , Pancreas/physiology , Polymerase Chain Reaction , Rats , Rats, Wistar
17.
Diabetes ; 55(3): 582-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16505219

ABSTRACT

The importance of mesenchymal-epithelial interactions in the proliferation of pancreatic progenitor cells is well established. Here, we provide evidence that the mesenchyme also controls the timing of beta-cell differentiation. When rat embryonic pancreatic epithelium was cultured without mesenchyme, we found first rapid induction in epithelial progenitor cells of the transcription factor neurogenin3 (Ngn3), a master gene controlling endocrine cell-fate decisions in progenitor cells; then beta-cell differentiation occurred. In the presence of mesenchyme, Ngn3 induction was delayed, and few beta-cells developed. This effect of the mesenchyme on Ngn3 induction was mediated by cell-cell contacts and required a functional Notch pathway. We then showed that associating Ngn3-expressing epithelial cells with mesenchyme resulted in poor beta-cell development via a mechanism mediated by soluble factors. Thus, in addition to its effect upstream of Ngn3, the mesenchyme regulated beta-cell differentiation downstream of Ngn3. In conclusion, these data indicate that the mesenchyme controls the timing of beta-cell differentiation both upstream and downstream of Ngn3.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/cytology , Mesoderm/physiology , Pancreas/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Communication , Male , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/physiology , Organ Culture Techniques , Rats , Rats, Wistar , Receptors, Notch/physiology , Signal Transduction
18.
Dev Dyn ; 234(2): 339-45, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16134081

ABSTRACT

In contrast to the considerable body of data on pancreas development in rodents, information on pancreas development in humans is scant. We previously described a model in which mature beta cells developed from human embryonic pancreas: human embryonic pancreas was grafted under the kidney capsule of scid mice, beta cells were then seen to develop in the graft. Here, we showed that not only beta cells, but also other endocrine cells, acinar cells and ducts develop in this model. We then used this model to probe the mechanisms underlying acinar and beta cell development in the human embryonic pancreas. BrdU pulse/chase experiments produced evidence of clonal acinar cell development: the first acinar cells to appear proliferated, thereby expanding the acinar cell population. In contrast, beta cell development was regulated by the proliferation of pancreatic progenitor cells, followed by beta-cell differentiation. We then showed that early progenitors expressing PDX1 proliferated, whereas late endocrine progenitors expressing Ngn3 did not. This proliferative capacity of early endocrine progenitor cells in embryonic human pancreas may hold promise for obtaining human beta-cell expansion.


Subject(s)
Cell Transplantation/methods , Pancreas/embryology , Pancreas/metabolism , Animals , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endocrine System/physiology , Homeodomain Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Insulin-Secreting Cells/metabolism , Mice , Mice, SCID , Microscopy, Fluorescence , Stem Cells/cytology , Trans-Activators , Transplantation, Heterologous
19.
Diabetes ; 52(8): 2035-42, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12882920

ABSTRACT

Islets of Langerhans are micro-organs scattered throughout the pancreas that contain insulin-producing cells, called beta-cells. Although new light has been recently shed on beta-cell development, information on the phenotype and location of beta-stem cells remains scarce. Here, we provide evidence that beta-stem cells are slow-cycling cells located within and around the islets of Langerhans. First, using a bromodeoxyuridine (BrdU) pulse/chase approach, we detected BrdU-retaining cells in vivo in the islet area of rat pancreata. These cells were negative for endocrine markers but expressed Pdx1, a marker for pancreatic stem cells. Next, using an in vitro model that mimicked endocrine cell development, we found that BrdU-retaining cells were capable of differentiating into beta-cells. Taken together, these observations demonstrate that BrdU retention is a property of beta-stem cells.


Subject(s)
Islets of Langerhans/cytology , Stem Cells/cytology , Animals , Antimetabolites/pharmacokinetics , Bromodeoxyuridine/pharmacokinetics , Cell Cycle/physiology , Cell Differentiation/physiology , Female , In Vitro Techniques , Islets of Langerhans/physiology , Pregnancy , Rats , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...