3,757 results on '"NEUROG3"'
Search Results
2. Single-cell transcriptome analysis of NEUROG3+ cells during pancreatic endocrine differentiation with small molecules
- Author
-
Jin Li, Junru Chen, Xiaoyu Luo, Guangxiu Lu, and Ge Lin
- Subjects
Pancreatic endocrine cells ,hESCs ,DAPT + 4FS ,NEUROG3 ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract The efficiency of inducing human embryonic stem cells into NEUROG3+ pancreatic endocrine cells is a bottleneck in stem cell therapy for diabetes. To understand the cell properties and fate decisions during differentiation, we analyzed the modified induction method using single-cell transcriptome and found that DAPT combined with four factors (4FS): nicotinamide, dexamethasone, forskolin and Alk5 inhibitor II (DAPT + 4FS) increased the expression of NEUROG3 to approximately 34.3%. The increased NEUROG3+ cells were mainly concentrated in Insulin + Glucagon + (INS + GCG+) and SLAC18A1 + Chromogranin A+(SLAC18A1 + CHGA +) populations, indicating that the increased NEUROG3+ cells promoted the differentiation of pancreatic endocrine cells and enterochromaffin-like cells. Single-cell transcriptome analysis provided valuable clues for further screening of pancreatic endocrine cells and differentiation of pancreatic islet cells. The gene set enrichment analysis (GSEA) suggest that we can try to promote the expression of INS + GCG+ population by up-regulating G protein-coupled receptor (GPCR) and mitogen-activated protein kinase signals and down-regulating Wnt, NIK/NF-KappaB and cytokine-mediated signal pathways. We can also try to regulate GPCR signaling through PLCE1, so as to increase the proportion of NEUROG3+ cells in INS+GCG+ populations. To exclude non-pancreatic endocrine cells, ALCAMhigh CD9low could be used as a marker for endocrine populations, and ALCAMhigh CD9lowCDH1low could remove the SLC18A1 + CHGA+ population.
- Published
- 2023
- Full Text
- View/download PDF
3. Null mutations of NEUROG3 are associated with delayed-onset diabetes mellitus.
- Author
-
Solorzano-Vargas, R Sergio, Bjerknes, Matthew, Wang, Jiafang, Wu, S Vincent, Garcia-Careaga, Manuel G, Pitukcheewanont, Pisit, Cheng, Hazel, German, Michael S, Georgia, Senta, and Martín, Martín G
- Subjects
Pancreas ,Islets of Langerhans ,Humans ,Malabsorption Syndromes ,Diabetes Mellitus ,Diabetes Mellitus ,Type 1 ,Genetic Predisposition to Disease ,Nerve Tissue Proteins ,Cell Proliferation ,Gene Expression Regulation ,Helix-Loop-Helix Motifs ,Child ,Female ,Male ,Enteroendocrine Cells ,Insulin-Secreting Cells ,Basic Helix-Loop-Helix Transcription Factors ,Promoter Regions ,Genetic ,Loss of Function Mutation ,Beta cells ,Diabetes ,Endocrinology ,Gastroenterology ,Islet cells ,Genetics ,Pediatric ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine - Abstract
Biallelic mutations of the gene encoding the transcription factor NEUROG3 are associated with a rare disorder that presents in neonates as generalized malabsorption - due to a complete absence of enteroendocrine cells - followed, in early childhood or beyond, by insulin-dependent diabetes mellitus (IDDM). The commonly delayed onset of IDDM suggests a differential requirement for NEUROG3 in endocrine cell generation in the human pancreas versus the intestine. However, previously identified human mutations were hypomorphic and, hence, may have had residual function in pancreas. We report 2 patients with biallelic functionally null variants of the NEUROG3 gene who nonetheless did not present with IDDM during infancy but instead developed permanent IDDM during middle childhood ages. The variants showed no evidence of function in traditional promoter-based assays of NEUROG3 function and also failed to exhibit function in a variety of potentially novel in vitro and in vivo molecular assays designed to discern residual NEUROG3 function. These findings imply that, unlike in mice, pancreatic endocrine cell generation in humans is not entirely dependent on NEUROG3 expression and, hence, suggest the presence of unidentified redundant in vivo pathways in human pancreas capable of yielding β cell mass sufficient to maintain euglycemia until early childhood.
- Published
- 2020
4. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network
- Author
-
Schreiber, Valérie, Mercier, Reuben, Jiménez, Sara, Ye, Tao, García-Sánchez, Emmanuel, Klein, Annabelle, Meunier, Aline, Ghimire, Sabitri, Birck, Catherine, Jost, Bernard, de Lichtenberg, Kristian Honnens, Honoré, Christian, Serup, Palle, and Gradwohl, Gérard
- Published
- 2021
- Full Text
- View/download PDF
5. Reduced Neurog3 Gene Dosage Shifts Enteroendocrine Progenitor Towards Goblet Cell Lineage in the Mouse Intestine
- Author
-
Li, Hui Joyce, Ray, Subir K., Kucukural, Alper, Gradwohl, Gerard, and Leiter, Andrew B.
- Published
- 2021
- Full Text
- View/download PDF
6. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network
- Author
-
Valérie Schreiber, Reuben Mercier, Sara Jiménez, Tao Ye, Emmanuel García-Sánchez, Annabelle Klein, Aline Meunier, Sabitri Ghimire, Catherine Birck, Bernard Jost, Kristian Honnens de Lichtenberg, Christian Honoré, Palle Serup, and Gérard Gradwohl
- Subjects
NEUROG3 ,iPSC ,Islet progenitors ,CUT&RUN ,T2DM ,SNPs ,Internal medicine ,RC31-1245 - Abstract
Objective: Mice lacking the bHLH transcription factor (TF) Neurog3 do not form pancreatic islet cells, including insulin-secreting beta cells, the absence of which leads to diabetes. In humans, homozygous mutations of NEUROG3 manifest with neonatal or childhood diabetes. Despite this critical role in islet cell development, the precise function of and downstream genetic programs regulated directly by NEUROG3 remain elusive. Therefore, we mapped genome-wide NEUROG3 occupancy in human induced pluripotent stem cell (hiPSC)–derived endocrine progenitors and determined NEUROG3 dependency of associated genes to uncover direct targets. Methods: We generated a novel hiPSC line (NEUROG3-HA-P2A-Venus) where NEUROG3 is HA-tagged and fused to a self-cleaving fluorescent VENUS reporter. We used the CUT&RUN technique to map NEUROG3 occupancy and epigenetic marks in pancreatic endocrine progenitors (PEP) that were differentiated from this hiPSC line. We integrated NEUROG3 occupancy data with chromatin status and gene expression in PEPs as well as their NEUROG3-dependence. In addition, we investigated whether NEUROG3 binds type 2 diabetes mellitus (T2DM)–associated variants at the PEP stage. Results: CUT&RUN revealed a total of 863 NEUROG3 binding sites assigned to 1263 unique genes. NEUROG3 occupancy was found at promoters as well as at distant cis-regulatory elements that frequently overlapped within PEP active enhancers. De novo motif analyses defined a NEUROG3 consensus binding motif and suggested potential co-regulation of NEUROG3 target genes by FOXA or RFX transcription factors. We found that 22% of the genes downregulated in NEUROG3−/− PEPs, and 10% of genes enriched in NEUROG3-Venus positive endocrine cells were bound by NEUROG3 and thus likely to be directly regulated. NEUROG3 binds to 138 transcription factor genes, some with important roles in islet cell development or function, such as NEUROD1, PAX4, NKX2-2, SOX4, MLXIPL, LMX1B, RFX3, and NEUROG3 itself, and many others with unknown islet function. Unexpectedly, we uncovered that NEUROG3 targets genes critical for insulin secretion in beta cells (e.g., GCK, ABCC8/KCNJ11, CACNA1A, CHGA, SCG2, SLC30A8, and PCSK1). Thus, analysis of NEUROG3 occupancy suggests that the transient expression of NEUROG3 not only promotes islet destiny in uncommitted pancreatic progenitors, but could also initiate endocrine programs essential for beta cell function. Lastly, we identified eight T2DM risk SNPs within NEUROG3-bound regions. Conclusion: Mapping NEUROG3 genome occupancy in PEPs uncovered unexpectedly broad, direct control of the endocrine genes, raising novel hypotheses on how this master regulator controls islet and beta cell differentiation.
- Published
- 2021
- Full Text
- View/download PDF
7. Phosphorylation of NEUROG3 Links Endocrine Differentiation to the Cell Cycle in Pancreatic Progenitors
- Author
-
Krentz, Nicole AJ, van Hoof, Dennis, Li, Zhongmei, Watanabe, Akie, Tang, Mei, Nian, Cuilan, German, Michael S, and Lynn, Francis C
- Subjects
Stem Cell Research - Nonembryonic - Non-Human ,Digestive Diseases ,Pancreatic Cancer ,Cancer ,Diabetes ,Stem Cell Research ,Rare Diseases ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Basic Helix-Loop-Helix Transcription Factors ,Cell Cycle ,Cell Differentiation ,Endocrine Cells ,Gene Expression Regulation ,Developmental ,Humans ,Islets of Langerhans ,Mice ,Nerve Tissue Proteins ,Pancreas ,Phosphorylation ,Stem Cells ,CRISPR/Cas9 ,Cdkn1b ,G1 lengthening ,Kras ,Ngn3 ,Sox9-rTTA ,diabetes ,human embryonic stem cells ,insulin ,mouse ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
During pancreatic development, proliferating pancreatic progenitors activate the proendocrine transcription factor neurogenin 3 (NEUROG3), exit the cell cycle, and differentiate into islet cells. The mechanisms that direct robust NEUROG3 expression within a subset of progenitor cells control the size of the endocrine population. Here we demonstrate that NEUROG3 is phosphorylated within the nucleus on serine 183, which catalyzes its hyperphosphorylation and proteosomal degradation. During progression through the progenitor cell cycle, NEUROG3 phosphorylation is driven by the actions of cyclin-dependent kinases 2 and 4/6 at G1/S cell-cycle checkpoint. Using models of mouse and human pancreas development, we show that lengthening of the G1 phase of the pancreatic progenitor cell cycle is essential for proper induction of NEUROG3 and initiation of endocrine cell differentiation. In sum, these studies demonstrate that progenitor cell-cycle G1 lengthening, through its actions on stabilization of NEUROG3, is an essential variable in normal endocrine cell genesis.
- Published
- 2017
8. Sustained Neurog3 Expression in Hormone-Expressing Islet Cells Is Required for Endocrine Maturation and Function
- Author
-
Wang, Sui, Jensen, Jan N., Seymour, Philip A., Hsu, Wei, Dor, Yuval, Sander, Maike, Magnuson, Mark A., Serup, Palle, Gu, Guoqiang, and Melton, Douglas A.
- Published
- 2009
- Full Text
- View/download PDF
9. Neurog3 misexpression unravels mouse pancreatic ductal cell plasticity.
- Author
-
Andhira Vieira, Bastien Vergoni, Monica Courtney, Noémie Druelle, Elisabet Gjernes, Biljana Hadzic, Fabio Avolio, Tiziana Napolitano, Sergi Navarro Sanz, Ahmed Mansouri, and Patrick Collombat
- Subjects
Medicine ,Science - Abstract
In the context of type 1 diabetes research and the development of insulin-producing β-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 β-like cells were found to be functional. Based on these findings, we suggest that ductal cells could represent a renewable source of new β-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.
- Published
- 2018
- Full Text
- View/download PDF
10. Effect of NEUROG3 polymorphism rs144643855 on regional spontaneous brain activity in major depressive disorder.
- Author
-
Hou Z, Liu X, Jiang W, Hou Z, Yin Y, Xie C, Zhang H, Zhang H, Zhang Z, and Yuan Y
- Subjects
- Adult, Depressive Disorder, Major diagnostic imaging, Female, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Prefrontal Cortex diagnostic imaging, Anhedonia physiology, Basic Helix-Loop-Helix Transcription Factors genetics, Connectome, Depressive Disorder, Major genetics, Depressive Disorder, Major physiopathology, Nerve Tissue Proteins genetics, Prefrontal Cortex physiopathology
- Abstract
Purpose: Our previous study identified a significant association between a single nucleotide polymorphism (SNP) located in the neurogenin3 (NEUROG3) gene and post-stroke depression (PSD) in Chinese populations. The present work explores whether polymorphism rs144643855 affects regional brain activity and clinical phenotypes in major depressive disorder (MDD)., Method: A total of 182 participants were included: 116 MDD patients and 66 normal controls. All participants underwent resting-state functional magnetic resonance imaging (rs-fMRI) scanning at baseline. Spontaneous brain activity was assessed using amplitude of low-frequency fluctuation (ALFF). The Hamilton Depression Scale-24 (HAMD-24) and Snaith-Hamilton Pleasure Scale (SHAPS) were used to assess participants at baseline. Two-way analysis of covariance (ANCOVA) was used to explore the interaction between diagnostic groups and NEUROG3 rs144643855 on regional brain activity. We performed correlation analysis to further test the association between these interactive brain regions and clinical manifestations of MDD., Results: Genotype and disease significantly interacted in the left inferior frontal gyrus (IFG-L), right superior frontal gyrus (SFG-R), and left paracentral lobule (PCL-L) (P < 0.05). ALFF values of the IFG-L were found to be significantly associated with anhedonia in MDD patients., Conclusion: These findings suggest a potential relationship between rs144643855 variations and altered frontal brain activity in MDD. NEUROG3 may play an important role in the neuropathophysiology of MDD., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Supplementary data for novel variants and phenotypes in NEUROG3 associated syndrome
- Author
-
Wejaphikul, Karn, Srilanchakon, Khomsak, Kamolvisit, Wuttichart, Jantasuwan, Supavadee, Santawong, Kanokwan, Tongkobpetch, Siraprapa, Theerapanon, Thanakorn, Damrongmanee, Alisara, Hongsawong, Nattaphorn, Ukarapol, Nuthapong, Dejkhamron, Prapai, Supornsilchai, Vichit, Porntaveetus, Thantrira, and Shotelersuk, Vorasuk
- Subjects
insulin ,diabetes ,enteroendocrine ,pancreas ,intestine ,neurogenin - Abstract
Supplementary data for novel variants and phenotypes in NEUROG3 associated syndrome.
- Published
- 2022
- Full Text
- View/download PDF
12. Bicaudal C1 promotes pancreatic NEUROG3+ endocrine progenitor differentiation and ductal morphogenesis
- Author
-
Lemaire, Laurence A, Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B, Grapin-Botton, Anne, Lemaire, Laurence A, Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B, and Grapin-Botton, Anne
- Abstract
In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.
- Published
- 2015
13. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network
- Subjects
Type 2 diabetes -- Genetic aspects ,Physical fitness ,Health - Abstract
2021 MAY 8 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- According to news reporting based on a preprint abstract, our journalists obtained [...]
- Published
- 2021
14. Genetic evidence that Nkx2.2 acts primarily downstream of Neurog3 in pancreatic endocrine lineage development
- Author
-
Angela J Churchill, Giselle Dominguez Gutiérrez, Ruth A Singer, David S Lorberbaum, Kevin A Fischer, and Lori Sussel
- Subjects
Transcriptional networks ,pancreatic islet ,beta cells ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Many pancreatic transcription factors that are essential for islet cell differentiation have been well characterized; however, because they are often expressed in several different cell populations, their functional hierarchy remains unclear. To parse out the spatiotemporal regulation of islet cell differentiation, we used a Neurog3-Cre allele to ablate Nkx2.2, one of the earliest and most broadly expressed islet transcription factors, specifically in the Neurog3+ endocrine progenitor lineage (Nkx2.2△endo). Remarkably, many essential components of the β cell transcriptional network that were down-regulated in the Nkx2.2KO mice, were maintained in the Nkx2.2△endo mice - yet the Nkx2.2△endo mice displayed defective β cell differentiation and recapitulated the Nkx2.2KO phenotype. This suggests that Nkx2.2 is not only required in the early pancreatic progenitors, but has additional essential activities within the endocrine progenitor population. Consistently, we demonstrate Nkx2.2 functions as an integral component of a modular regulatory program to correctly specify pancreatic islet cell fates.
- Published
- 2017
- Full Text
- View/download PDF
15. Novel variants and phenotypes in NEUROG3 associated syndrome
- Author
-
Karn Wejaphikul, Khomsak Srilanchakon, Wuttichart Kamolvisit, Supavadee Jantasuwan, Kanokwan Santawong, Siraprapa Tongkobpetch, Thanakorn Theerapanon, Alisara Damrongmanee, Nattaphorn Hongsawong, Nuthapong Ukarapol, Prapai Dejkhamron, Vichit Supornsilchai, Thantrira Porntaveetus, and Vorasuk Shotelersuk
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Biochemistry (medical) ,Clinical Biochemistry ,Biochemistry - Abstract
Context Biallelic pathogenic variants in the NEUROG3 gene cause malabsorptive diarrhea, insulin-dependent diabetes mellitus (IDDM), and rarely hypogonadotropic hypogonadism. With only 17 reported cases, the clinical and mutational spectra of this disease are far from complete. Objective To identify the underlying genetic etiology in 3 unrelated Thai patients who presented with early-onset malabsorptive diarrhea, endocrine abnormalities, and renal defects and to determine the pathogenicity of the newly identified pathogenic variants using luciferase reporter assays and western blot. Methods Three unrelated patients with congenital diarrhea were recruited. Detailed clinical and endocrinological features were obtained. Exome sequencing was performed to identify mutations and in vitro functional experiments including luciferase reporter assay were studied to validate their pathogenicity. Results In addition to malabsorptive diarrhea due to enteric anendocrinosis, IDDM, short stature, and delayed puberty, our patients also exhibited pituitary gland hypoplasia with multiple pituitary hormone deficiencies (Patient 1, 2, 3) and proximal renal tubulopathy (Patient 2, 3) that have not previously reported. Exome sequencing revealed that Patient 1 was homozygous for c.371C > G (p.Thr124Arg) while the other 2 patients were homozygous for c.284G > C (p.Arg95Pro) in NEUROG3. Both variants have never been previously reported. Luciferase reporter assay demonstrated that these 2 variants impaired transcriptional activity of NEUROG3. Conclusions This study reported pituitary gland hypoplasia with multiple pituitary hormone deficiencies and proximal renal tubulopathy and 2 newly identified NEUROG3 loss-of-function variants in the patients with NEUROG3-associated syndrome.
- Published
- 2022
16. Bicaudal C1 promotes pancreatic NEUROG3+ endocrine progenitor differentiation and ductal morphogenesis.
- Author
-
UCL - SSS/DDUV - Institut de Duve, Lemaire, Laurence A, Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B, Grapin-Botton, Anne, UCL - SSS/DDUV - Institut de Duve, Lemaire, Laurence A, Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B, and Grapin-Botton, Anne
- Abstract
In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.
- Published
- 2015
17. The Basic Helix-Loop-Helix Transcription Factor NEUROG3 Is Required for Development of the Human Endocrine Pancreas.
- Author
-
McGrath, Patrick S., Watson, Carey L., Ingram, Cameron, Helmrath, Michael A., and Wells, James M.
- Subjects
HELIX-loop-helix motifs ,MICROPHTHALMIA-associated transcription factor ,TRANSCRIPTION factors ,PANCREATIC physiology ,ENDOCRINE system physiology - Abstract
Neurogenin3 (NEUROG3) is a basic helix-loop-helix transcription factor required for development of the endocrine pancreas in mice. In contrast, humans with NEUROG3 mutations are born with endocrine pancreas function, calling into question whether NEUROG3 is required for human endocrine pancreas development. To test this directly, we generated human embryonic stem cell (hESC) lines where both alleles of NEUROG3 were disrupted using CRISPR/Cas9-mediated gene targeting. NEUROG3
-/- hESC lines efficiently formed pancreatic progenitors but lacked detectible NEUROG3 protein and did not form endocrine cells in vitro. Moreover, NEUROG3-/- hESC lines were unable to form mature pancreatic endocrine cells after engraftment of PDX1+ /NKX6.1+ pancreatic progenitors into mice. In contrast, a 75-90% knockdown of NEUROG3 caused a reduction, but not a loss, of pancreatic endocrine cell development. We conclude that NEUROG3 is essential for endocrine pancreas development in humans and that as little as 10% NEUROG3 is sufficient for formation of pancreatic endocrine cells. [ABSTRACT FROM AUTHOR]- Published
- 2015
- Full Text
- View/download PDF
18. Bicaudal C1 promotes pancreatic NEUROG3(+) endocrine progenitor differentiation and ductal morphogenesis (vol 142, pg 858, 2015)
- Author
-
Lemaire, Laurence A., Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B., Grapin-Botton, Anne, Lemaire, Laurence A., Goulley, Joan, Kim, Yung Hae, Carat, Solenne, Jacquemin, Patrick, Rougemont, Jacques, Constam, Daniel B., and Grapin-Botton, Anne
- Published
- 2016
19. The transcriptional activity of Neurog3 affects migration and differentiation of ectopic endocrine cells in chicken endoderm.
- Author
-
Rosenberg, Louise C., Lafon, Merete L., Pedersen, Jesper Karup, Yassin, Hani, Jensen, Jan Nygaard, Serup, Palle, and Hecksher-Sørensen, Jacob
- Abstract
Neurog3 is expressed transiently in pancreatic endocrine progenitors where it is responsible for activating a transcription factor cascade which eventually defines the mature endocrine cells. However, the mechanism by which Neurog3 regulates different aspects of the endocrine differentiation program is less clear. In this report we used in ovo electroporation to investigate how manipulation of Neurog3 protein activity affected migration, differentiation and fate determination. We found that changes in the onset of Neurog3 expression only had minor effect on differentiation. However increasing the transcriptional activity of Neurog3 by fusing it to VP16 or co-electroporating with Ep300 caused the electroporated cells to migrate rather than differentiate. In contrast, reducing the transcriptional activity of Neurog3 by deleting parts of the activation domain, by fusing Neurog3 to the engrailed repressor domain, or co-electroporating with Hdac1 greatly increased the proportion of glucagon expressing cells. Developmental Dynamics 239:1950-1966, 2010. © 2010 Wiley-Liss, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
20. A novel NEUROG3 mutation in neonatal diabetes associated with a neuro‐intestinal syndrome.
- Author
-
Hancili, Suna, Bonnefond, Amélie, Philippe, Julien, Vaillant, Emmanuel, De Graeve, Franck, Sand, Olivier, Busiah, Kanetee, Robert, Jean‐Jacques, Polak, Michel, Froguel, Philippe, Güven, Ayla, and Vaxillaire, Martine
- Subjects
- *
GENETICS of diabetes , *INTESTINAL diseases , *GENES , *GENETIC mutation , *TRANSCRIPTION factors , *SEQUENCE analysis , *CHILDREN , *GENETICS - Abstract
Neonatal diabetes mellitus (NDM) is a rare form of non‐autoimmune diabetes usually diagnosed in the first 6 months of life. Various genetic defects have been shown to cause NDM with diverse clinical presentations and variable severity. Among transcriptional factor genes associated with isolated or syndromic NDM, a few cases of homozygous mutations in the NEUROG3 gene have been reported, all mutated patients presenting with congenital malabsorptive diarrhea with or without diabetes at a variable age of onset from early life to childhood. Through a targeted next‐generation sequencing assay for monogenic diabetes genes, we aimed to search for pathogenic deleterious mutation in a Turkish patient with NDM, severe malabsorptive diarrhea, neurointestinal dysplasia and other atypical features. In this patient, we identified a novel homozygous nonsense mutation (p.Q4*) in NEUROG3. The same biallelic mutation was found in another affected family member. Of note, the study proband presents with abnormalities of the intrahepatic biliary tract, thyroid gland and central nervous system, which has never been reported before in NEUROG3 mutation carriers. Our findings extend the usually described clinical features associated with NEUROG3 deficiency in humans, and question the extent to which a complete lack of NEUROG3 expression may affect pancreas endocrine function in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
21. NEUROG3 is a critical downstream effector for STAT3-regulated differentiation of mammalian stem and progenitor spermatogonia.
- Author
-
Kaucher, Amy V and Kaucher, Amy V
- Abstract
Spermatogenesis relies on coordinated differentiation of stem and progenitor spermatogonia, and the transcription factor STAT3 is essential for this process in mammals. Here we studied the THY1+ spermatogonial population in mouse testes, which contains spermatogonial stem cells (SSC) and non-stem cell progenitor spermatogonia, to further define the downstream mechanism regulating differentiation. Transcript abundance for the bHLH transcription factor Neurog3 was found to be significantly reduced upon transient inhibition of STAT3 signaling in these cells and exposure to GDNF, a key growth factor regulating self-renewal of SSCs, suppressed activation of STAT3 and in accordance Neurog3 gene expression. Moreover, STAT3 was found to bind the distal Neurog3 promoter/enhancer region in THY1+ spermatogonia and regulate transcription. Transient inhibition of Neurog3 expression in cultures of proliferating THY1+ spermatogonia increased stem cell content after several self-renewal cycles without effecting overall proliferation of the cells, indicating impaired differentiation of SSCs to produce progenitor spermatogonia. Furthermore, cultured THY1+ spermatogonia with induced deficiency of Neurog3 were found to be incapable of differentiation in vivo following transplantation into testes of recipient mice. Collectively, these results establish a mechanism by which activation of STAT3 regulates the expression of NEUROG3 to subsequently drive differentiation of SSC and progenitor spermatogonia in the mammalian germline.
- Published
- 2012
22. Severe NEUROG3 mutation underlies pancreatic endocrine and exocrine insufficiency
- Author
-
Senta Georgia, Kyle R. Vogt, Martin G. Martin, Cambrian Liu, Katelyn Millette, Juliana Austin, Yuhua Zheng, and Shengmei Zhou
- Subjects
business.industry ,Mutation (genetic algorithm) ,Cancer research ,Endocrine system ,Medicine ,business - Abstract
Patients with NEUROG3 mutations suffer from diabetes mellitus due to pancreatic endocrinopathy and chronic malabsorptive diarrhea due to enteric endocrinopathy. We have identified a severe truncation mutation in NEUROG3 (P39PfsX38) that results in pancreatic exocrine insufficiency and significantly contributes to chronic malabsorptive diarrhea. We identified this novel phenotype by interrogating induced pluripotent stem cells from the NEUROG3-P39PfsX38 patient’s fibroblasts and an isogenic “wild-type” control cell line generated via CRISPR-Cas9 gene editing. We discovered that NEUROG3- P39PfsX38 lines failed to activate pancreatic progenitor and differentiated lineage markers, suggesting that the mutation may disrupt pancreatic organogenesis and could result in endocrine and exocrine dysfunction. Isogenic corrected cell lines differentiated into all pancreatic lineages. Clinical assessments concluded that the patient has exocrine pancreatic insufficiency. Treatment with pancreatic enzyme replacement therapy improved patient outcomes, including weight gain, fat absorption, and resolution of fat-soluble vitamin deficiency. These results expose a novel role for NEUROG3 in human pancreatic differentiation and illustrate how patient-specific stem cells can be used to interrogate disease etiology and affect patient care.
- Published
- 2021
23. Single-cell transcriptome and accessible chromatin dynamics during endocrine pancreas development
- Author
-
Duvall, Eliza, Benitez, Cecil M, Tellez, Krissie, Enge, Martin, Pauerstein, Philip T, Li, Lingyu, Baek, Songjoon, Quake, Stephen R, Smith, Jason P, Sheffield, Nathan C, Kim, Seung K, and Arda, H Efsun
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Stem Cell Research - Nonembryonic - Non-Human ,Cancer ,Orphan Drug ,Pancreatic Cancer ,Rare Diseases ,Human Genome ,Digestive Diseases ,Diabetes ,Biotechnology ,Stem Cell Research ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Metabolic and endocrine ,Animals ,Basic Helix-Loop-Helix Transcription Factors ,Cell Differentiation ,Cell Lineage ,Chromatin ,Gene Expression Regulation ,Developmental ,Islets of Langerhans ,Mice ,Nerve Tissue Proteins ,Single-Cell Analysis ,Transcriptome ,ATAC-seq ,Neurog3 ,pancreas ,scRNA-seq - Abstract
Delineating gene regulatory networks that orchestrate cell-type specification is a continuing challenge for developmental biologists. Single-cell analyses offer opportunities to address these challenges and accelerate discovery of rare cell lineage relationships and mechanisms underlying hierarchical lineage decisions. Here, we describe the molecular analysis of mouse pancreatic endocrine cell differentiation using single-cell transcriptomics, chromatin accessibility assays coupled to genetic labeling, and cytometry-based cell purification. We uncover transcription factor networks that delineate β-, α-, and δ-cell lineages. Through genomic footprint analysis, we identify transcription factor-regulatory DNA interactions governing pancreatic cell development at unprecedented resolution. Our analysis suggests that the transcription factor Neurog3 may act as a pioneer transcription factor to specify the pancreatic endocrine lineage. These findings could improve protocols to generate replacement endocrine cells from renewable sources, like stem cells, for diabetes therapy.
- Published
- 2022
24. Severe NEUROG3 mutation underlies pancreatic endocrine and exocrine insufficiency
- Author
-
Georgia, Senta, primary, Millette, Katelyn, additional, Zheng, Yuhua, additional, Vogt, Kyle, additional, Liu, Cambrian, additional, Austin, Juliana, additional, Zhou, Shengmei, additional, and Martin, Martin, additional
- Published
- 2021
- Full Text
- View/download PDF
25. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network
- Author
-
Schreiber, Valérie, primary, Mercier, Reuben, additional, Jiménez, Sara, additional, Ye, Tao, additional, García-Sánchez, Emmanuel, additional, Klein, Annabelle, additional, Meunier, Aline, additional, Ghimire, Sabitri, additional, Birck, Catherine, additional, Jost, Bernard, additional, de Lichtenberg, Kristian Honnens, additional, Honoré, Christian, additional, Serup, Palle, additional, and Gradwohl, Gérard, additional
- Published
- 2021
- Full Text
- View/download PDF
26. Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium
- Author
-
Gerbe, F., van Es, J.H., Makrini, L., Brulin, B., Mellitzer, G., Robine, S., Romagnolo, B., Shroyer, N.F., Bourgaux, J.F., Pignodel, C., Clevers, H., Jay, P., Gerbe, F., van Es, J.H., Makrini, L., Brulin, B., Mellitzer, G., Robine, S., Romagnolo, B., Shroyer, N.F., Bourgaux, J.F., Pignodel, C., Clevers, H., and Jay, P.
- Abstract
The unique morphology of tuft cells was first revealed by electron microscopy analyses in several endoderm-derived epithelia. Here, we explore the relationship of these cells with the other cell types of the intestinal epithelium and describe the first marker signature allowing their unambiguous identification. We demonstrate that although mature tuft cells express DCLK1, a putative marker of quiescent stem cells, they are post-mitotic, short lived, derive from Lgr5-expressing epithelial stem cells, and are found in mouse and human tumors. We show that whereas the ATOH1/MATH1 transcription factor is essential for their differentiation, Neurog3, SOX9, GFI1, and SPDEF are dispensable, which distinguishes these cells from enteroendocrine, Paneth, and goblet cells, and raises from three to four the number of secretory cell types in the intestinal epithelium. Moreover, we show that tuft cells are the main source of endogenous intestinal opioids and are the only epithelial cells that express cyclooxygenase enzymes, suggesting important roles for these cells in the intestinal epithelium physiopathology., The unique morphology of tuft cells was first revealed by electron microscopy analyses in several endoderm-derived epithelia. Here, we explore the relationship of these cells with the other cell types of the intestinal epithelium and describe the first marker signature allowing their unambiguous identification. We demonstrate that although mature tuft cells express DCLK1, a putative marker of quiescent stem cells, they are post-mitotic, short lived, derive from Lgr5-expressing epithelial stem cells, and are found in mouse and human tumors. We show that whereas the ATOH1/MATH1 transcription factor is essential for their differentiation, Neurog3, SOX9, GFI1, and SPDEF are dispensable, which distinguishes these cells from enteroendocrine, Paneth, and goblet cells, and raises from three to four the number of secretory cell types in the intestinal epithelium. Moreover, we show that tuft cells are the main source of endogenous intestinal opioids and are the only epithelial cells that express cyclooxygenase enzymes, suggesting important roles for these cells in the intestinal epithelium physiopathology.
- Published
- 2011
27. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas
- Author
-
Wang, Sui, Yan, Jingbo, Anderson, Daniel A., Xu, Yanwen, Kanal, Maneesh C., Cao, Zheng, Wright, Christopher V.E., and Gu, Guoqiang
- Subjects
Developmental biology ,Diabetes ,Mice ,Biological sciences - Abstract
To link to full-text access for this article, visit this link: http://dx.doi.org/10.1016/j.ydbio.2009.12.009 Byline: Sui Wang, Jingbo Yan, Daniel A. Anderson, Yanwen Xu, Maneesh C. Kanal, Zheng Cao, Christopher V.E. Wright, Guoqiang Gu Keywords: Diabetes; Islet; Neurog3; Ngn3; Lateral inhibition; Notch; Endocrine progenitor; Dosage; Haploinsufficiency; Pancreas Abstract: The basic helix-loop-helix transcription factor Neurog3 (Neurogenin3 or Ngn3) actively drives endodermal progenitor cells towards endocrine islet cell differentiation during embryogenesis. Here, we manipulate Neurog3 expression levels in endocrine progenitor cells without altering its expression pattern using heterozygosity and a hypomorph. Lowered Neurog3 gene dosage in the developing pancreatic epithelium reduces the overall production of endocrine islet cells without significantly affecting the proportions of various islet cell types that do form. A reduced Neurog3 production level in the endocrine-directed pancreatic progenitor population activates the expression of Neurog3 in an increased number of epithelial progenitors. Yet a significant number of these Neurog3.sup.+ cells detected in heterozygous and hypomorphic pancreata, possibly those that express low levels of Neurog3, move on to adopt pancreatic ductal or acinar fates. These data directly demonstrate that achieving high levels of Neurog3 expression is a critical step for endocrine commitment from multipotent pancreatic progenitors. These findings also suggest that a high level of Neurog3 expression could mediate lateral inhibition or other unknown feedback mechanisms to regulate the number of cells that initiate Neurog3 transcription and protein production. The control of Neurog3.sup.+ cell number and the Neurog3 threshold-dependent endocrine differentiation mechanism combine to select a specific proportion of pancreatic progenitor cells to adopt the islet cell fate. Author Affiliation: Program in Developmental Biology and Department of Cell and Developmental Biology, Center for Stem Cell Biology, Vanderbilt University Medical Center, 465 21st Avenue South, Rm 4128, Vanderbilt Medical Center, Nashville, TN 37232, USA Article History: Received 1 July 2009; Revised 5 December 2009; Accepted 7 December 2009
- Published
- 2010
28. Null mutations of NEUROG3 are associated with delayed-onset diabetes mellitus
- Author
-
R. Sergio Solorzano-Vargas, Senta Georgia, Jiafang Wang, Manuel Garcia-Careaga, Matthew Bjerknes, Martin G. Martin, Michael S. German, Pisit Pitukcheewanont, S. Vincent Wu, and Hazel Cheng
- Subjects
0301 basic medicine ,Male ,Malabsorption ,Enteroendocrine cell ,0302 clinical medicine ,Endocrinology ,Loss of Function Mutation ,Insulin-Secreting Cells ,Basic Helix-Loop-Helix Transcription Factors ,2.1 Biological and endogenous factors ,Aetiology ,Promoter Regions, Genetic ,Child ,Pediatric ,Helix-Loop-Helix Motifs ,Diabetes ,Gastroenterology ,Islet cells ,General Medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Pancreas ,Research Article ,Type 1 ,endocrine system ,medicine.medical_specialty ,Enteroendocrine Cells ,Nerve Tissue Proteins ,Biology ,Promoter Regions ,03 medical and health sciences ,Islets of Langerhans ,Malabsorption Syndromes ,Genetic ,In vivo ,Diabetes mellitus ,Internal medicine ,medicine ,Diabetes Mellitus ,Genetics ,Humans ,Genetic Predisposition to Disease ,Gene ,Transcription factor ,Metabolic and endocrine ,Cell Proliferation ,Beta cells ,medicine.disease ,In vitro ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Gene Expression Regulation - Abstract
Biallelic mutations of the gene encoding the transcription factor NEUROG3 are associated with a rare disorder that presents in neonates as generalized malabsorption - due to a complete absence of enteroendocrine cells - followed, in early childhood or beyond, by insulin-dependent diabetes mellitus (IDDM). The commonly delayed onset of IDDM suggests a differential requirement for NEUROG3 in endocrine cell generation in the human pancreas versus the intestine. However, previously identified human mutations were hypomorphic and, hence, may have had residual function in pancreas. We report 2 patients with biallelic functionally null variants of the NEUROG3 gene who nonetheless did not present with IDDM during infancy but instead developed permanent IDDM during middle childhood ages. The variants showed no evidence of function in traditional promoter-based assays of NEUROG3 function and also failed to exhibit function in a variety of potentially novel in vitro and in vivo molecular assays designed to discern residual NEUROG3 function. These findings imply that, unlike in mice, pancreatic endocrine cell generation in humans is not entirely dependent on NEUROG3 expression and, hence, suggest the presence of unidentified redundant in vivo pathways in human pancreas capable of yielding β cell mass sufficient to maintain euglycemia until early childhood.
- Published
- 2020
29. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas
- Author
-
Christopher V.E. Wright, Daniel A. Anderson, Maneesh C. Kanal, Jingbo Yan, Zheng Cao, Sui Wang, Guoqiang Gu, and Yanwen Xu
- Subjects
Islet ,endocrine system ,medicine.medical_specialty ,Cell type ,Notch ,Cellular differentiation ,Endocrine progenitor ,Population ,Gene Dosage ,Neurog3 ,Mice, Transgenic ,Nerve Tissue Proteins ,Haploinsufficiency ,Cell fate determination ,Biology ,Article ,Ngn3 ,Mice ,03 medical and health sciences ,Dosage ,Exocrine Glands ,0302 clinical medicine ,Endocrine Glands ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Lateral inhibition ,Cell Lineage ,Progenitor cell ,education ,Pancreas ,Molecular Biology ,030304 developmental biology ,Progenitor ,0303 health sciences ,geography ,education.field_of_study ,geography.geographical_feature_category ,Reverse Transcriptase Polymerase Chain Reaction ,Diabetes ,Cell Biology ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The basic helix–loop–helix transcription factor Neurog3 (Neurogenin3 or Ngn3) actively drives endodermal progenitor cells towards endocrine islet cell differentiation during embryogenesis. Here, we manipulate Neurog3 expression levels in endocrine progenitor cells without altering its expression pattern using heterozygosity and a hypomorph. Lowered Neurog3 gene dosage in the developing pancreatic epithelium reduces the overall production of endocrine islet cells without significantly affecting the proportions of various islet cell types that do form. A reduced Neurog3 production level in the endocrine-directed pancreatic progenitor population activates the expression of Neurog3 in an increased number of epithelial progenitors. Yet a significant number of these Neurog3+ cells detected in heterozygous and hypomorphic pancreata, possibly those that express low levels of Neurog3, move on to adopt pancreatic ductal or acinar fates. These data directly demonstrate that achieving high levels of Neurog3 expression is a critical step for endocrine commitment from multipotent pancreatic progenitors. These findings also suggest that a high level of Neurog3 expression could mediate lateral inhibition or other unknown feedback mechanisms to regulate the number of cells that initiate Neurog3 transcription and protein production. The control of Neurog3+ cell number and the Neurog3 threshold-dependent endocrine differentiation mechanism combine to select a specific proportion of pancreatic progenitor cells to adopt the islet cell fate.
- Published
- 2010
30. Novel Variants and Phenotypes in NEUROG3-Associated Syndrome.
- Author
-
Wejaphikul, Karn, Srilanchakon, Khomsak, Kamolvisit, Wuttichart, Jantasuwan, Supavadee, Santawong, Kanokwan, Tongkobpetch, Siraprapa, Theerapanon, Thanakorn, Damrongmanee, Alisara, Hongsawong, Nattaphorn, Ukarapol, Nuthapong, Dejkhamron, Prapai, Supornsilchai, Vichit, Porntaveetus, Thantrira, and Shotelersuk, Vorasuk
- Subjects
PHENOTYPES ,DIABETES ,HYPOGONADISM - Abstract
Context: Biallelic pathogenic variants in the NEUROG3 gene cause malabsorptive diarrhea, insulin-dependent diabetes mellitus (IDDM), and rarely hypogonadotropic hypogonadism. With only 17 reported cases, the clinical and mutational spectra of this disease are far from complete. Objective: To identify the underlying genetic etiology in 3 unrelated Thai patients who presented with early-onset malabsorptive diarrhea, endocrine abnormalities, and renal defects and to determine the pathogenicity of the newly identified pathogenic variants using luciferase reporter assays and western blot. Methods: Three unrelated patients with congenital diarrhea were recruited. Detailed clinical and endocrinological features were obtained. Exome sequencing was performed to identify mutations and in vitro functional experiments including luciferase reporter assay were studied to validate their pathogenicity. Results: In addition to malabsorptive diarrhea due to enteric anendocrinosis, IDDM, short stature, and delayed puberty, our patients also exhibited pituitary gland hypoplasia with multiple pituitary hormone deficiencies (Patient 1, 2, 3) and proximal renal tubulopathy (Patient 2, 3) that have not previously reported. Exome sequencing revealed that Patient 1 was homozygous for c.371C>G (p.Thr124Arg) while the other 2 patients were homozygous for c.284G>C (p.Arg95Pro) in NEUROG3. Both variants have never been previously reported. Luciferase reporter assay demonstrated that these 2 variants impaired transcriptional activity of NEUROG3. Conclusions: This study reported pituitary gland hypoplasia with multiple pituitary hormone deficiencies and proximal renal tubulopathy and 2 newly identified NEUROG3 loss-of-function variants in the patients with NEUROG3-associated syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
31. Permanent Neonatal Diabetes and Enteric Anendocrinosis Associated With Biallelic Mutations in NEUROG3
- Author
-
Rubio-Cabezas, Oscar, Jensen, Jan N., Hodgson, Maria I., Codner, Ethel, Ellard, Sian, Serup, Palle, and Hattersley, Andrew T.
- Published
- 2011
- Full Text
- View/download PDF
32. Exposure to PM2.5 during pregnancy or lactation increases methylation while reducing the expression of Pdx1 and NEUROG3 in mouse pancreatic islets
- Author
-
Maria José de Carvalho Costa, Guilherme Francisco Peruca, Patrícia O. Prada, Kellen Cristina da Cruz Rodrigues, Daisuke Hayashi Neto, Gabriel Forato Anhê, Paulo Hilário Nascimento Saldiva, Victor Yuji Yariwake, Raquel Patrícia Ataíde Lima, Junia Carolina Santos-Silva, Vitor Ferreira Boico, and Mariana Matera Veras
- Subjects
Andrology ,endocrine system ,medicine.anatomical_structure ,Lactation ,Pancreatic islets ,DNA methylation ,medicine ,PDX1 ,Glucose homeostasis ,Promoter ,Methylation ,Epigenetics ,Biology - Abstract
Air pollution is comprised of several substances, including particulate matter (PM). Exposure to air pollution may trigger alterations in DNA methylation thus modifying gene expression patterns. This phenomenon is likely to mediate the relationship between exposure to air pollution and adverse health effects. The purpose of this study was analyzing the effects of exposure to PM2.5 during pregnancy or lactation and whether it would cause multigenerational epigenetic alterations in the promoter region of the genes Pdx1 and NEUROG3 within mouse pancreatic islets. Our results show that maternal exposure to PM2.5 led to an elevation in blood glucose levels within the two following generations (F1 and F2). There was also an increase in DNA methylation in the aforementioned promoter regions accompanied by reduced gene expression in generations F1 and F2 upon F0 exposure to PM2.5 during pregnancy. These data suggest that maternal exposure to PM2.5 from air pollution, particularly during pregnancy, may lead to a multigenerational and lifelong negative impact on glucose homeostasis mediated by an increase in DNA methylation within the promoter region of the genes Pdx1 and NEUROG3 in pancreatic islets.
- Published
- 2019
- Full Text
- View/download PDF
33. ROCK-nmMyoII, Notch and Neurog3 gene-dosage link epithelial morphogenesis with cell fate in the pancreatic endocrine-progenitor niche.
- Author
-
Bankaitis ED, Bechard ME, Gu G, Magnuson MA, and Wright CVE
- Subjects
- Animals, Cell Differentiation genetics, Cell Movement, Mice, Mice, Transgenic, Pancreas cytology, Stem Cells cytology, Transcriptional Activation genetics, Basic Helix-Loop-Helix Transcription Factors genetics, Endocrine Cells cytology, Gene Dosage genetics, Nerve Tissue Proteins genetics, Organogenesis genetics, Pancreas embryology, Receptors, Notch genetics, rho-Associated Kinases genetics
- Abstract
During mouse pancreas organogenesis, endocrine cells are born from progenitors residing in an epithelial plexus niche. After a period in a lineage-primed Neurog3
LO state, progenitors become endocrine committed via upregulation of Neurog3 We find that the Neurog3LO transcriptional upregulation still occur without Neurog3 protein, suggesting that morphogenetic cues deployed within the plexus initiate endocrine commitment upstream or independently of Neurog3. Neurog3 is required for cell-rear detachment and complete endocrine-cell birth. The ROCK-nmMyoII pathway coordinates epithelial-cell morphogenesis and the progression through Neurog3HI transition is associated with distinct stages of an epithelial egression process: narrowing the apical surface of the cell, basalward cell movement and eventual cell-rear detachment from the apical lumen surface to allow clustering as nascent islets under the basement membrane. Apical narrowing, basalward movement and Neurog3 transcriptional upregulation still occur without Neurog3 protein, suggesting that morphogenetic cues deployed within the plexus initiate endocrine commitment upstream or independently of Neurog3. Neurog3 is required for cell-rear detachment and complete endocrine-cell birth. The ROCK-nmMyoII pathway coordinates epithelial-cell morphogenesis and the progression through Neurog3 -expressing states. NmMyoII is necessary for apical narrowing, basalward cell displacement and Neurog3 upregulation, but all three are limited by ROCK activity. We propose that ROCK-nmMyoII activity, Neurog3 gene-dose and Notch signaling integrate endocrine fate allocation with epithelial plexus growth and morphogenesis, representing a feedback control circuit that coordinates morphogenesis with lineage diversification in the endocrine-birth niche., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)- Published
- 2018
- Full Text
- View/download PDF
34. Neurog3 misexpression unravels mouse pancreatic ductal cell plasticity.
- Author
-
Vieira A, Vergoni B, Courtney M, Druelle N, Gjernes E, Hadzic B, Avolio F, Napolitano T, Navarro Sanz S, Mansouri A, and Collombat P
- Subjects
- 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, 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
- Abstract
In the context of type 1 diabetes research and the development of insulin-producing β-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 β-like cells were found to be functional. Based on these findings, we suggest that ductal cells could represent a renewable source of new β-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., Competing Interests: Author MC was not employed by Evotec at the time of this study; however, she is currently employed by Evotec International GmbH. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2018
- Full Text
- View/download PDF
35. Exposure to PM2.5 during pregnancy or lactation increases methylation while reducing the expression of Pdx1 and NEUROG3 in mouse pancreatic islets
- Author
-
Ataíde Lima, Raquel Patricia, primary, Boico, Vitor Ferreira, additional, Peruca, Guilherme Francisco, additional, Cruz Rodrigues, Kellen Cristina da, additional, Yariwake, Victor Yuji, additional, Neto, Daisuke Hayashi, additional, de Carvalho Costa, Maria José, additional, Rebelo Santos-Silva, Junia Carolina, additional, Anhê, Gabriel Forato, additional, Nascimento Saldiva, Paulo Hilário, additional, Veras, Mariana Matera, additional, and de Oliveira Prada, Patrícia, additional
- Published
- 2019
- Full Text
- View/download PDF
36. Mutations in IAPP and NEUROG3 genes are not a common cause of permanent neonatal/infancy/childhood-onset diabetes
- Author
-
Nocerino, V., Colombo, C., Bonfanti, R., Iafusco, D., and Barbetti, F.
- Published
- 2009
- Full Text
- View/download PDF
37. Mutations in the Coding Region of the Neurogenin 3 Gene (NEUROG3) Are Not a Common Cause of Maturity-Onset Diabetes of the Young in Japanese Subjects
- Author
-
del Bosque-Plata, Laura, Lin, Joseph, Horikawa, Yukio, Schwarz, Peter E.H., Cox, Nancy J., Iwasaki, Naoko, Ogata, Makiko, Iwamoto, Yasuhiko, German, Michael S., and Bell, Graeme I.
- Published
- 2001
38. Newly identified congenital malabsorptive diarrhea is caused by mutations in NEUROG3
- Published
- 2006
39. ROCK-nmMyoII, Notch, and Neurog3 gene-dosage link epithelial morphogenesis with cell fate in the pancreatic endocrine-progenitor niche
- Author
-
Christopher V.E. Wright, Guoqiang Gu, Matthew E. Bechard, Mark A. Magnuson, and Eric D. Bankaitis
- Subjects
Transcriptional Activation ,0301 basic medicine ,endocrine system ,Organogenesis ,Gene Dosage ,Notch signaling pathway ,Morphogenesis ,Mice, Transgenic ,Nerve Tissue Proteins ,Enteroendocrine cell ,Biology ,Cell fate determination ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Cell Movement ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Progenitor cell ,Pancreas ,Molecular Biology ,Progenitor ,rho-Associated Kinases ,Receptors, Notch ,Stem Cells ,Cell Differentiation ,Cell biology ,030104 developmental biology ,Endocrine Cells ,Research Article ,Developmental Biology - Abstract
During mouse pancreas organogenesis, endocrine cells are born from progenitors residing in an epithelial plexus niche. After a period in a lineage-primed Neurog3LO state, progenitors become endocrine committed via upregulation of Neurog3. We find that the Neurog3LO to Neurog3HI transition is associated with distinct stages of an epithelial egression process: narrowing the apical surface of the cell, basalward cell movement and eventual cell-rear detachment from the apical lumen surface to allow clustering as nascent islets under the basement membrane. Apical narrowing, basalward movement and Neurog3 transcriptional upregulation still occur without Neurog3 protein, suggesting that morphogenetic cues deployed within the plexus initiate endocrine commitment upstream or independently of Neurog3. Neurog3 is required for cell-rear detachment and complete endocrine-cell birth. The ROCK-nmMyoII pathway coordinates epithelial-cell morphogenesis and the progression through Neurog3-expressing states. NmMyoII is necessary for apical narrowing, basalward cell displacement and Neurog3 upregulation, but all three are limited by ROCK activity. We propose that ROCK-nmMyoII activity, Neurog3 gene-dose and Notch signaling integrate endocrine fate allocation with epithelial plexus growth and morphogenesis, representing a feedback control circuit that coordinates morphogenesis with lineage diversification in the endocrine-birth niche.
- Published
- 2018
40. Neurog3 misexpression unravels mouse pancreatic ductal cell plasticity
- Author
-
Biljana Hadzic, Bastien Vergoni, Andhira Vieira, Elisabet Gjernes, Monica Courtney, Tiziana Napolitano, Sergi Navarro Sanz, Patrick Collombat, Fabio Avolio, Ahmed Mansouri, and Noémie Druelle
- Subjects
Male ,0301 basic medicine ,Cell Plasticity ,Test Statistics ,lcsh:Medicine ,Enteroendocrine cell ,Biochemistry ,Epithelium ,Mice ,Endocrinology ,Mathematical and Statistical Techniques ,Animal Cells ,Insulin-Secreting Cells ,Medicine and Health Sciences ,Basic Helix-Loop-Helix Transcription Factors ,Insulin ,lcsh:Science ,Multidisciplinary ,geography.geographical_feature_category ,Genetically Modified Organisms ,Animal Models ,Islet ,Cell biology ,medicine.anatomical_structure ,Experimental Organism Systems ,Physical Sciences ,Cellular Types ,Anatomy ,Genetic Engineering ,Pancreas ,Immunohistochemical Analysis ,Statistics (Mathematics) ,Research Article ,Biotechnology ,Ductal cells ,Transgene ,Endocrine System ,Mouse Models ,Mice, Transgenic ,Nerve Tissue Proteins ,Context (language use) ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Exocrine Glands ,medicine ,Animals ,Humans ,Regeneration ,Statistical Methods ,Immunohistochemistry Techniques ,Diabetic Endocrinology ,geography ,Genetically Modified Animals ,Regeneration (biology) ,lcsh:R ,Pancreatic Ducts ,Biology and Life Sciences ,Cell Biology ,Hormones ,Histochemistry and Cytochemistry Techniques ,Disease Models, Animal ,Biological Tissue ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Immunologic Techniques ,lcsh:Q ,Endocrine Cells ,Mathematics - Abstract
In the context of type 1 diabetes research and the development of insulin-producing β-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 β-like cells were found to be functional. Based on these findings, we suggest that ductal cells could represent a renewable source of new β-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.
- Published
- 2018
41. Revisiting the immunocytochemical detection of Neurogenin 3 expression in mouse and man.
- Author
-
Honoré, C., Rescan, C., Hald, J., McGrath, P. S., Petersen, M. B. K., Hansson, M., Klein, T., Østergaard, S., Wells, J. M., and Madsen, O. D.
- Subjects
NEUROGENIN 3 ,PROGENITOR cells ,PANCREATIC beta cells ,NEUROGENINS ,ISLANDS of Langerhans - Abstract
During embryonic development, endocrine cells of the pancreas are specified from multipotent progenitors. The transcription factor Neurogenin 3 ( NEUROG3) is critical for this development and it has been shown that all endocrine cells of the pancreas arise from endocrine progenitors expressing NEUROG3. A thorough understanding of the role of NEUROG3 during development, directed differentiation of pluripotent stem cells and in models of cellular reprogramming, will guide future efforts directed at finding novel sources of β-cells for cell replacement therapies. In this article, we review the expression and function of NEUROG3 in both mouse and human and present the further characterization of a monoclonal antibody directed against NEUROG3. This antibody has been previously been used for detection of both mouse and human NEUROG3. However, our results suggest that the epitope recognized by this antibody is specific to mouse NEUROG3. Thus, we have also generated a monoclonal antibody specifically recognizing human NEUROG3 and present the characterization of this antibody here. Together, these antibodies will provide useful tools for future studies of NEUROG3 expression, and the data presented in this article suggest that recently described expression patterns of NEUROG3 in human foetal and adult pancreas should be re-examined. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
42. FUCCI tracking shows cell-cycle-dependent Neurog3 variation in pancreatic progenitors.
- Author
-
Bechard ME, Bankaitis ED, Ustione A, Piston DW, Magnuson MA, and Wright CVE
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Cells, Cultured, Embryonic Stem Cells cytology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Islets of Langerhans cytology, Mice, Nerve Tissue Proteins metabolism, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Cycle, Embryonic Stem Cells metabolism, Islets of Langerhans metabolism, Nerve Tissue Proteins genetics
- Abstract
During pancreas organogenesis, Neurog3
HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO ). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI ) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2 -M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment., (© 2017 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
43. Genetic evidence that Nkx2.2 acts primarily downstream of Neurog3 in pancreatic endocrine lineage development.
- Author
-
Churchill AJ, Gutiérrez GD, Singer RA, Lorberbaum DS, Fischer KA, and Sussel L
- Subjects
- Animals, Homeobox Protein Nkx-2.2, Mice, Mice, Knockout, Zebrafish Proteins, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Differentiation, Homeodomain Proteins metabolism, Insulin-Secreting Cells physiology, Islets of Langerhans cytology, Nerve Tissue Proteins metabolism, Transcription Factors metabolism
- Abstract
Many pancreatic transcription factors that are essential for islet cell differentiation have been well characterized; however, because they are often expressed in several different cell populations, their functional hierarchy remains unclear. To parse out the spatiotemporal regulation of islet cell differentiation, we used a Neurog3-Cre allele to ablate Nkx2.2 , one of the earliest and most broadly expressed islet transcription factors, specifically in the Neurog3
+ endocrine progenitor lineage ( Nkx2.2△endo ). Remarkably, many essential components of the β cell transcriptional network that were down-regulated in the Nkx2.2KO mice, were maintained in the Nkx2.2△endo mice - yet the Nkx2.2△endo mice displayed defective β cell differentiation and recapitulated the Nkx2.2KO phenotype. This suggests that Nkx2.2 is not only required in the early pancreatic progenitors, but has additional essential activities within the endocrine progenitor population. Consistently, we demonstrate Nkx2.2 functions as an integral component of a modular regulatory program to correctly specify pancreatic islet cell fates., Competing Interests: The authors declare that no competing interests exist.- Published
- 2017
- Full Text
- View/download PDF
44. The Basic Helix-Loop-Helix Transcription Factor NEUROG3 Is Required for Development of the Human Endocrine Pancreas
- Author
-
James M. Wells, Patrick S. McGrath, Cameron Ingram, Carey L. Watson, and Michael A. Helmrath
- Subjects
endocrine system ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Molecular Sequence Data ,Nerve Tissue Proteins ,Enteroendocrine cell ,Biology ,Islets of Langerhans ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,Internal Medicine ,medicine ,Humans ,Endocrine system ,Progenitor cell ,Transcription factor ,Cells, Cultured ,Embryonic Stem Cells ,Base Sequence ,Gene targeting ,Embryonic stem cell ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Islet Studies ,Mutation ,PDX1 ,Pancreas - Abstract
Neurogenin3 (NEUROG3) is a basic helix-loop-helix transcription factor required for development of the endocrine pancreas in mice. In contrast, humans with NEUROG3 mutations are born with endocrine pancreas function, calling into question whether NEUROG3 is required for human endocrine pancreas development. To test this directly, we generated human embryonic stem cell (hESC) lines where both alleles of NEUROG3 were disrupted using CRISPR/Cas9-mediated gene targeting. NEUROG3−/− hESC lines efficiently formed pancreatic progenitors but lacked detectible NEUROG3 protein and did not form endocrine cells in vitro. Moreover, NEUROG3−/− hESC lines were unable to form mature pancreatic endocrine cells after engraftment of PDX1+/NKX6.1+ pancreatic progenitors into mice. In contrast, a 75–90% knockdown of NEUROG3 caused a reduction, but not a loss, of pancreatic endocrine cell development. We conclude that NEUROG3 is essential for endocrine pancreas development in humans and that as little as 10% NEUROG3 is sufficient for formation of pancreatic endocrine cells.
- Published
- 2015
45. Researchers from University of Massachusetts Report on Findings in Gastroenterology and Hepatology (Reduced Neurog3 Gene Dosage Shifts Enteroendocrine Progenitor Towards Goblet Cell Lineage In the Mouse Intestine)
- Subjects
United States. National Institutes of Health -- Reports ,United States. National Human Genome Research Institute -- Reports ,Genetic research -- Reports ,Health ,University of Massachusetts -- Reports - Abstract
2021 APR 5 (NewsRx) -- By a News Reporter-Staff News Editor at Gastroenterology Week -- Investigators discuss new findings in Health and Medicine - Gastroenterology and Hepatology. According to news [...]
- Published
- 2021
46. Precommitment low-level Neurog3 expression defines a long-lived mitotic endocrine-biased progenitor pool that drives production of endocrine-committed cells.
- Author
-
Bechard ME, Bankaitis ED, Hipkens SB, Ustione A, Piston DW, Yang YP, Magnuson MA, and Wright CV
- Subjects
- Animals, Cell Differentiation, Cell Proliferation genetics, Mice, Mitosis, Pancreas cytology, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Endocrine Cells cytology, Gene Expression Regulation, Developmental, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Stem Cells cytology
- Abstract
The current model for endocrine cell specification in the pancreas invokes high-level production of the transcription factor Neurogenin 3 (Neurog3) in Sox9(+) bipotent epithelial cells as the trigger for endocrine commitment, cell cycle exit, and rapid delamination toward proto-islet clusters. This model posits a transient Neurog3 expression state and short epithelial residence period. We show, however, that a Neurog3(TA.LO) cell population, defined as Neurog3 transcriptionally active and Sox9(+) and often containing nonimmunodetectable Neurog3 protein, has a relatively high mitotic index and prolonged epithelial residency. We propose that this endocrine-biased mitotic progenitor state is functionally separated from a pro-ductal pool and endows them with long-term capacity to make endocrine fate-directed progeny. A novel BAC transgenic Neurog3 reporter detected two types of mitotic behavior in Sox9(+) Neurog3(TA.LO) progenitors, associated with progenitor pool maintenance or derivation of endocrine-committed Neurog3(HI) cells, respectively. Moreover, limiting Neurog3 expression dramatically increased the proportional representation of Sox9(+) Neurog3(TA.LO) progenitors, with a doubling of its mitotic index relative to normal Neurog3 expression, suggesting that low Neurog3 expression is a defining feature of this cycling endocrine-biased state. We propose that Sox9(+) Neurog3(TA.LO) endocrine-biased progenitors feed production of Neurog3(HI) endocrine-committed cells during pancreas organogenesis., (© 2016 Bechard et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2016
- Full Text
- View/download PDF
47. FUCCI tracking shows cell-cycle-dependent Neurog3 variation in pancreatic progenitors
- Author
-
Eric D. Bankaitis, Mark A. Magnuson, Alessandro Ustione, Christopher V.E. Wright, Matthew E. Bechard, and David W. Piston
- Subjects
0301 basic medicine ,endocrine system ,Population ,Green Fluorescent Proteins ,Organogenesis ,Nerve Tissue Proteins ,SOX9 ,Biology ,Article ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Endocrinology ,Genetics ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Progenitor cell ,education ,Mitosis ,Cells, Cultured ,Embryonic Stem Cells ,Progenitor ,education.field_of_study ,Cell Cycle ,Cell Biology ,Cell cycle ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Pancreas - Abstract
During pancreas organogenesis, Neurog3HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO ). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI ) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2 -M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment.
- Published
- 2017
48. FUCCI tracking shows that Neurog3 levels vary with cell-cycle phase in endocrine-biased pancreatic progenitors
- Author
-
Eric D. Bankaitis, Christopher V.E. Wright, Matthew E. Bechard, Alessandro Ustione, David W. Piston, and Mark A. Magnuson
- Subjects
Genetics ,endocrine system ,0303 health sciences ,education.field_of_study ,Transcriptional activity ,Population ,SOX9 ,Biology ,Cell cycle phase ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Endocrine system ,Progenitor cell ,education ,Mitosis ,030217 neurology & neurosurgery ,030304 developmental biology ,Progenitor - Abstract
Neurog3HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2-M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment.
- Published
- 2017
49. A novel NEUROG3 mutation in neonatal diabetes associated with a neuro-intestinal syndrome
- Author
-
Ayla Güven, Emmanuel Vaillant, Amélie Bonnefond, Franck De Graeve, J. Philippe, Suna Hancili, Philippe Froguel, Olivier Sand, Martine Vaxillaire, Jean-Jacques Robert, Kanetee Busiah, and Michel Polak
- Subjects
0301 basic medicine ,Proband ,Biallelic Mutation ,Male ,endocrine system ,Endocrinology, Diabetes and Metabolism ,Nonsense mutation ,030209 endocrinology & metabolism ,Nerve Tissue Proteins ,Bioinformatics ,Diabetes Complications ,03 medical and health sciences ,0302 clinical medicine ,Neonatal diabetes mellitus ,Malabsorption Syndromes ,Diabetes mellitus ,Internal Medicine ,Basic Helix-Loop-Helix Transcription Factors ,Medicine ,Humans ,Child ,Genetics ,business.industry ,medicine.disease ,030104 developmental biology ,Dysplasia ,Codon, Nonsense ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Mutation (genetic algorithm) ,Female ,Age of onset ,business - Abstract
Neonatal diabetes mellitus (NDM) is a rare form of non-autoimmune diabetes usually diagnosed in the first 6 months of life. Various genetic defects have been shown to cause NDM with diverse clinical presentations and variable severity. Among transcriptional factor genes associated with isolated or syndromic NDM, a few cases of homozygous mutations in the NEUROG3 gene have been reported, all mutated patients presenting with congenital malabsorptive diarrhea with or without diabetes at a variable age of onset from early life to childhood. Through a targeted next-generation sequencing assay for monogenic diabetes genes, we aimed to search for pathogenic deleterious mutation in a Turkish patient with NDM, severe malabsorptive diarrhea, neurointestinal dysplasia and other atypical features. In this patient, we identified a novel homozygous nonsense mutation (p.Q4*) in NEUROG3. The same biallelic mutation was found in another affected family member. Of note, the study proband presents with abnormalities of the intrahepatic biliary tract, thyroid gland and central nervous system, which has never been reported before in NEUROG3 mutation carriers. Our findings extend the usually described clinical features associated with NEUROG3 deficiency in humans, and question the extent to which a complete lack of NEUROG3 expression may affect pancreas endocrine function in humans.
- Published
- 2017
50. Insm1 promotes endocrine cell differentiation by modulating the expression of a network of genes that includes Neurog3 and Ripply3
- Author
-
Shinji Takada, Anna B. Osipovich, Tadashi Okubo, Qiaoming Long, Elisabetta Manduchi, Rama Gangula, Judsen Schneider, Christian J. Stoeckert, Mark A. Magnuson, and Susan B. Hipkens
- Subjects
Time Factors ,Mouse ,Transcription, Genetic ,Cellular differentiation ,Enteroendocrine cell ,Cell Separation ,Mice ,0302 clinical medicine ,Cell Movement ,Genes, Reporter ,Basic Helix-Loop-Helix Transcription Factors ,Pancreas development ,Gene Regulatory Networks ,Mice, Knockout ,0303 health sciences ,Stem Cells ,Gene Expression Regulation, Developmental ,Cell migration ,Cell Differentiation ,Stem Cells and Regeneration ,Flow Cytometry ,Cell biology ,Extracellular Matrix ,DNA-Binding Proteins ,medicine.anatomical_structure ,PDX1 ,Stem cell ,Pancreas ,RNA Splicing ,Green Fluorescent Proteins ,Nerve Tissue Proteins ,Biology ,03 medical and health sciences ,medicine ,Transcription factors ,Animals ,Cell Lineage ,Progenitor cell ,Molecular Biology ,Alleles ,030304 developmental biology ,Cell Proliferation ,Cell growth ,Repressor Proteins ,Alternative Splicing ,RNA ,Gene expression ,Endocrine progenitor cells ,Endocrine Cells ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Insulinoma associated 1 (Insm1) plays an important role in regulating the development of cells in the central and peripheral nervous systems, olfactory epithelium and endocrine pancreas. To better define the role of Insm1 in pancreatic endocrine cell development we generated mice with an Insm1GFPCre reporter allele and used them to study Insm1-expressing and null populations. Endocrine progenitor cells lacking Insm1 were less differentiated and exhibited broad defects in hormone production, cell proliferation and cell migration. Embryos lacking Insm1 contained greater amounts of a non-coding Neurog3 mRNA splice variant and had fewer Neurog3/Insm1 co-expressing progenitor cells, suggesting that Insm1 positively regulates Neurog3. Moreover, endocrine progenitor cells that express either high or low levels of Pdx1, and thus may be biased towards the formation of specific cell lineages, exhibited cell type-specific differences in the genes regulated by Insm1. Analysis of the function of Ripply3, an Insm1-regulated gene enriched in the Pdx1-high cell population, revealed that it negatively regulates the proliferation of early endocrine cells. Taken together, these findings indicate that in developing pancreatic endocrine cells Insm1 promotes the transition from a ductal progenitor to a committed endocrine cell by repressing a progenitor cell program and activating genes essential for RNA splicing, cell migration, controlled cellular proliferation, vasculogenesis, extracellular matrix and hormone secretion.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.