Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
1.
Acta Diabetol ; 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39190183

RÉSUMÉ

AIMS: Monogenic diabetes is one of the few examples in metabolic diseases in which a real precision medicine approach can be implemented in daily clinical work. Unfortunately, most of what is known today comes from studies in Whites, thus leaving much uncertainty about the genetics and the clinical presentation of monogenic diabetes in non-Europeans. To fill this gap, we report here two pedigrees from Bangladesh with CEL- and RFX6- diabetes, two rare types of monogenic diabetes which have never been described so far in individuals of the Indian subcontinent. METHODS: Next generation, Sanger sequencing and Multiplex Ligation-dependent Probe Amplification (MLPA) were performed. Variants' interpretation was according to the American College of Medical Genetics and Genomics guidelines. RESULTS: In the pedigree with CEL-diabetes, a large and never described deletion of exon 2-11 of CEL (confirmed by MLPA) affecting the entire catalytic domain and being likely pathogenic (LP) was observed in both the proband (who had diabetes at 16) and his mother (diabetes at 31), but not in relatives with normoglycemia. In the pedigree with RFX6-diabetes, a LP protein truncation variant (PTV, p.Tyr192*) in RFX6 was found in both the proband (diabetes at 9) and his mother (diabetes at 30), thus suggesting high heterogeneity in disease onset. Normoglycemic relatives were not available for genetic testing. CONCLUSIONS: We report genetic features and clinical presentation of the first two cases of CEL- and RFX6-diabetes from the Indian subcontinent, thus contributing to fill the gap of knowledge on monogenic diabetes in non-Europeans.

2.
Adv Sci (Weinh) ; 11(32): e2401492, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38932472

RÉSUMÉ

Genetic and epigenetic alterations are cancer hallmark characteristics. However, the role of inherited cancer predisposition alleles in co-opting lineage factor epigenetic reprogramming and tumor progression remains elusive. Here the FinnGen cohort phenome-wide analysis, along with multiple genome-wide association studies, has consistently identified the rs339331-RFX6/6q22 locus associated with prostate cancer (PCa) risk across diverse populations. It is uncovered that rs339331 resides in a reprogrammed androgen receptor (AR) binding site in PCa tumors, with the T risk allele enhancing AR chromatin occupancy. RFX6, an AR-regulated gene linked to rs339331, exhibits synergistic prognostic value for PCa recurrence and metastasis. This comprehensive in vitro and in vivo studies demonstrate the oncogenic functions of RFX6 in promoting PCa cell proliferation and metastasis. Mechanistically, RFX6 upregulates HOXA10 that profoundly correlates with adverse PCa outcomes and is pivotal in RFX6-mediated PCa progression, facilitating the epithelial-mesenchymal transition (EMT) and modulating the TGFß/SMAD signaling axis. Clinically, HOXA10 elevation is associated with increased EMT scores, tumor advancement and PCa recurrence. Remarkably, reducing RFX6 expression restores enzalutamide sensitivity in resistant PCa cells and tumors. This findings reveal a complex interplay of genetic and epigenetic mechanisms in PCa pathogenesis and drug resistance, centered around disrupted prostate lineage AR signaling and abnormal RFX6 expression.


Sujet(s)
Allèles , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Protéines à homéodomaine , Tumeurs de la prostate , Facteurs de transcription des facteurs régulateurs X , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Humains , Mâle , Souris , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Étude d'association pangénomique/méthodes , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Facteurs de transcription des facteurs régulateurs X/génétique , Facteurs de transcription des facteurs régulateurs X/métabolisme , Transduction du signal/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique
3.
Development ; 151(9)2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38587174

RÉSUMÉ

The gastrointestinal (GI) tract is complex and consists of multiple organs with unique functions. Rare gene variants can cause congenital malformations of the human GI tract, although the molecular basis of these has been poorly studied. We identified a patient with compound-heterozygous variants in RFX6 presenting with duodenal malrotation and atresia, implicating RFX6 in development of the proximal intestine. To identify how mutations in RFX6 impact intestinal patterning and function, we derived induced pluripotent stem cells from this patient to generate human intestinal organoids (HIOs). We identified that the duodenal HIOs and human tissues had mixed regional identity, with gastric and ileal features. CRISPR-mediated correction of RFX6 restored duodenal identity. We then used gain- and loss-of-function and transcriptomic approaches in HIOs and Xenopus embryos to identify that PDX1 is a downstream transcriptional target of RFX6 required for duodenal development. However, RFX6 had additional PDX1-independent transcriptional targets involving multiple components of signaling pathways that are required for establishing early regional identity in the GI tract. In summary, we have identified RFX6 as a key regulator in intestinal patterning that acts by regulating transcriptional and signaling pathways.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Protéines à homéodomaine , Organoïdes , Facteurs de transcription des facteurs régulateurs X , Transactivateurs , Humains , Facteurs de transcription des facteurs régulateurs X/génétique , Facteurs de transcription des facteurs régulateurs X/métabolisme , Animaux , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique , Organoïdes/métabolisme , Organoïdes/embryologie , Duodénum/métabolisme , Duodénum/embryologie , Intestins/embryologie , Atrésie intestinale/génétique , Cellules souches pluripotentes induites/métabolisme , Plan d'organisation du corps/génétique , Transduction du signal/génétique , Mutation/génétique
4.
Clin Transl Med ; 13(12): e1511, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-38093528

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) cells undergo reprogramming of glucose metabolism to support uncontrolled proliferation, of which the intrinsic mechanism still merits further investigation. Although regulatory factor X6 (RFX6) is aberrantly expressed in different cancers, its precise role in cancer development remains ambiguous. METHODS: Microarrays of HCC tissues were employed to investigate the expression of RFX6 in tumour and adjacent non-neoplastic tissues. Functional assays were employed to explore the role of RFX6 in HCC development. Chromatin immunoprecipitation, untargeted metabolome profiling and sequencing were performed to identify potential downstream genes and pathways regulated by RFX6. Metabolic assays were employed to investigate the effect of RFX6 on glycolysis in HCC cells. Bioinformatics databases were used to validate the above findings. RESULTS: HCC tissues exhibited elevated expression of RFX6. High RFX6 expression represented as an independent hazard factor correlated to poor prognosis in patients with HCC. RFX6 deficiency inhibited HCC development in vitro and in vivo, while its overexpression exerted opposite functions. Mechanistically, RFX6 bound to the promoter area of phosphoglycerate mutase 1 (PGAM1) and upregulated its expression. The increased PGAM1 protein levels enhanced glycolysis and further promoted the development of HCC. CONCLUSIONS: RFX6 acted as a novel driver for HCC development by promoting aerobic glycolysis, disclosing the potential of the RFX6-PGAM1 axis for therapeutic targeting.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/métabolisme , Prolifération cellulaire/génétique , Glycolyse/génétique , Tumeurs du foie/métabolisme , Phosphoglyceromutase/génétique , Phosphoglyceromutase/métabolisme
5.
Article de Anglais | MEDLINE | ID: mdl-38054414

RÉSUMÉ

Objective: Maturity-onset diabetes of the young (MODY) is the most common type of monogenic diabetes. To date, mutations have been identified in 14 different genes of patients with a clinical diagnosis of MODY. This study screened mutations in 14 MODY-related genes and the regulator factor X6 (RFX6) gene in children. Materials and Methods: The presence of clinical features of MODY and negative results for three autoantibody markers of T1DM in children and adolescents were used as inclusion criteria for genetic testing. The screening panel for next-generation sequencing included 14 MODY-related genes (GCK, HNF4A, HNF1A, HNF1B, PDX1, NEUROD1, KLF11, CEL, PAX4, INS, BLK, ABCC8, KCNJ11, and APPL1) and the RFX6 gene. Results: Twenty-four different variants in MODY-related genes were identified in 49 children diagnosed with autoantibody-negative type 1 diabetes mellitus (T1DM). A 12 variants were classified as P/LP while 12 were interpreted as variant of unknown significance (VUS). Nine of the pathogenic or likely pathogenic variants were found in GCK, two in HNF1B, and one in ABCC8. Three variants were novel, and one was a de novo variant. All of the variants, except one, showed heterozygotic inheritance. Conclusion: This study screened mutations in the 14 MODY-related genes and the regulatory factor X6 (RFX6) gene in Turkish children diagnosed with autoantibody-negative type 1 diabetes mellitus (T1DM). The frequencies of the MODY subtypes differed from previous reports. Although GCK-MODY was the most frequent mutation in Turkish children, similar to previous studies, the second most prevalent MODY subtype was HNF1B-MODY. This study also established three additional novel mutations in different MODY genes.

6.
Front Endocrinol (Lausanne) ; 13: 802351, 2022.
Article de Anglais | MEDLINE | ID: mdl-35813646

RÉSUMÉ

Aims/Hypothesis: Caused by biallelic mutations of the gene encoding the transcription factor RFX6, the rare Mitchell-Riley syndrome (MRS) comprises neonatal diabetes, pancreatic hypoplasia, gallbladder agenesis or hypoplasia, duodenal atresia, and severe chronic diarrhea. So far, sixteen cases have been reported, all with a poor prognosis. This study discusses the multidisciplinary intensive clinical management of 4 new cases of MRS that survived over the first 2 years of life. Moreover, it demonstrates how the mutations impair the RFX6 function. Methods: Clinical records were analyzed and described in detail. The functional impact of two RFX6R181W and RFX6V506G variants was assessed by measuring their ability to transactivate insulin transcription and genes that encode the L-type calcium channels required for normal pancreatic beta-cell function. Results: All four patients were small for gestational age (SGA) and prenatally diagnosed with duodenal atresia. They presented with neonatal diabetes early in life and were treated with intravenous insulin therapy before switching to subcutaneous insulin pump therapy. All patients faced recurrent hypoglycemic episodes, exacerbated when parenteral nutrition (PN) was disconnected. A sensor-augmented insulin pump therapy with a predictive low-glucose suspension system was installed with good results. One patient had a homozygous c.1517T>G (p.Val506Gly) mutation, two patients had a homozygous p.Arg181Trp mutation, and one patient presented with new compound heterozygosity. The RFX6V506G and RFX6R181W mutations failed to transactivate the expression of insulin and genes that encode L-type calcium channel subunits required for normal pancreatic beta-cell function. Conclusions/Interpretation: Multidisciplinary and intensive disease management improved the clinical outcomes in four patients with MRS, including adjustment of parenteral/oral nutrition progression and advanced diabetes technologies. A better understanding of RFX6 function, in both intestine and pancreas cells, may break ground in new therapies, particularly regarding the use of drugs that modulate the enteroendocrine system.


Sujet(s)
Diabète , Maladies néonatales , Diabète/diagnostic , Occlusion duodénale , Maladies de la vésicule biliaire , Humains , Nouveau-né , Insuline/génétique , Atrésie intestinale , Mutation , Facteurs de transcription des facteurs régulateurs X/génétique , Facteurs de transcription des facteurs régulateurs X/métabolisme
7.
Pediatr Transplant ; 26(5): e14270, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35307919

RÉSUMÉ

BACKGROUND: MRS/MFS is a rare multisystem disorder with a poor prognosis. The high mortality rate of this syndrome is related to the severity of the associated gastrointestinal, pancreatic, and hepatobiliary conditions, as most of them are not amenable to conventional medical and surgical treatments. METHODS: We report the case of a Romani girl with all the key clinical features of MRS/MFS, and a review of cases reported in the literature. Our patient is a newborn from consanguineous parents who presented duodenal atresia, hypoplastic pancreas, gallbladder agenesis, and neonatal diabetes. Given the clinical suspicion of MRS/MFS, a genetic analysis was performed which revealed the presence of a homozygous variant in the RFX6 gene. During the course of the disease, the patient presented intractable secretory diarrhea and severe intestinal failure. RESULTS: At 2 years of age, she underwent MVT of the stomach, duodenum, small intestine, colon, liver, and pancreas. There were no surgical complications. Histologic evaluation of the small bowel showed extensive patches of gastric heterotopia. After more than 10 years of follow-up, she had presented with normal gastrointestinal, hepatic, and pancreatic function. She has one of the longest survival periods in the literature. CONCLUSIONS: Our experience suggests that multivisceral transplantation may be a promising option in select cases of MRS/MFS.


Sujet(s)
Diabète , Maladies de la vésicule biliaire , Atrésie intestinale , Diabète/génétique , Femelle , Maladies de la vésicule biliaire/génétique , Maladies de la vésicule biliaire/anatomopathologie , Humains , Nouveau-né , Atrésie intestinale/génétique , Atrésie intestinale/anatomopathologie , Atrésie intestinale/chirurgie , Fistule trachéo-oesophagienne
8.
Article de Anglais | MEDLINE | ID: mdl-35331396

RÉSUMÉ

Congenital enteropathies (CE) are a group of rare inherited diseases with a typical onset early in life. They involve defects in enterocyte structure or differentiation. They can cause a severe condition of intestinal failure (IF). The diagnostic approach is based first on clinical presentation (consanguinity, prenatal expression, polyhydramnios, early neonatal onset, aspect of stools, persistence at bowel rest, associated extra-digestive manifestations….) and histo-pathological analyses. These rare intestinal diseases cause protracted diarrhea that might resolve, for a few, with a dietetic approach. However, protracted or permanent IF may require long term parenteral nutrition and, in limited cases, intestinal transplantation. With the progresses in both clinical nutrition and genetics, many of these CE are nowadays associated with recognized gene mutations. It improved our knowledge and the understanding in the patho-physiology of these diseases, thus, leading potentially to therapeutic perspectives. These review cover most of the early onset CE and excludes the immune related diarrhea.


Sujet(s)
Entérocytes , Maladies intestinales , Diarrhée/étiologie , Diarrhée/thérapie , Entérocytes/anatomopathologie , Humains , Nouveau-né , Maladies intestinales/diagnostic , Maladies intestinales/génétique , Maladies intestinales/thérapie , Intestins/anatomopathologie , Nutrition parentérale
9.
Orphanet J Rare Dis ; 16(1): 455, 2021 10 29.
Article de Anglais | MEDLINE | ID: mdl-34715892

RÉSUMÉ

BACKGROUND: Homozygous mutations in the transcription factor RFX6 are the cause of the Mitchell-Riley syndrome (MRS) associating neonatal diabetes, congenital digestive system, such as biliary atresia, pancreatic hypoplasia, duodenal and/or jejunal atresia, intestinal malrotation, gallbladder aplasia, cholestasis. A constitutive inactivation of RFX6 leads also to gastric heterotopia. Application of RNA-seq in human diseases may help to better understand pathogenic mechanism of diseases and to predict the risk of developing chronic disorders and personalizing their prevention and treatment. We evaluated oncogenic patterns and cancer predisposition using the transcriptomic profile in a case of MRS with neonatal diabetes, duodenal atresia, and extensive intestinal tract gastric heterotopia. RESULTS: We signalled the interactors of RFX6 with other up and downregulated genes, that may be interested in severity of diabetic condition, in multi-organs impairment and cancer predisposition. Furthermore, several dysregulated genes are involved in biological processes that can lead to promote cancer including "Evading apoptosis" (BAD, BBC3, EGF, FGFR2, FLT3LG, HMOX1, HRAS, IFNAR2, IGF1R, IL12RB1, IL13RA1, IL15, IL2RB, IL2RG, IL6R, KEAP1, MGST1, PDGFA, PDGFRB, PIK3R3, RALB, RALGDS, RASSF1, SOS1, TGFA, TXNRD3), "Proliferation" (APC, BRAF, CCND2, CCND3, CCNE2, FGFR2, FLT3LG, FZD1, FZD6, HMOX1, HRAS, IGF1R, KEAP1, LRP6, MAPK3, MGST1, PDGFA, PDGFB, PDGFRB, RB1, SOS1, TGFA, TXNRD3, WNT10B), "Sustained angiogenesis" (BRAF, FGFR2, FLT3LG, HRAS, IGF1R, JAG1, MAPK3, NOTCH2, PDGFA, PDGFB, PDGFRB, SOS1, TGFA, TGFB1), "Genomic instability" (BAD, BBC3) and "Insensitivity to anti-growth signals" (SMAD2, TGFB1). We also inspected the signalings and their related genes in cancer, such as "PI3K signaling", "ERK signaling", "JAK-STAT signaling", "Calcium signaling", "Other RAS signaling", "WNT signaling". CONCLUSIONS: In our MRS patient, we signaled the interactors of RFX6 with other up- and downregulated genes that may be related to severe diabetic condition, multi-organ impairment, and cancer predisposition. Notably, many dysregulated genes may lead to triggering carcinogenesis. The possibility of the patient developing cancer degeneration in heterotopic gastric mucosa and/or additional long-term tumoral sequelae is not excluded. Personalized prevention and treatment strategies should be proposed.


Sujet(s)
Diabète , Atrésie intestinale , Tumeurs , Carcinogenèse , Occlusion duodénale , Maladies de la vésicule biliaire , Muqueuse gastrique/métabolisme , Humains , Nouveau-né , Atrésie intestinale/génétique , Protéine-1 de type kelch associée à ECH , Facteur-2 apparenté à NF-E2 , Phosphatidylinositol 3-kinases , Facteurs de transcription des facteurs régulateurs X/génétique , Facteurs de transcription des facteurs régulateurs X/métabolisme , Transcriptome
10.
J Pediatr Endocrinol Metab ; 34(12): 1619-1622, 2021 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-34416793

RÉSUMÉ

OBJECTIVES: Monogenic diabetes mellitus (DM) is a single gene disorder, primarily characterized by impairment in the development or function of pancreatic beta cells. CASE PRESENTATION: A 14-year-old girl was initially diagnosed with type 2 DM. The patient did not have any anti-islet autoantibody and showed acanthosis nigricans. She was managed with long-acting insulin and oral hypoglycemic agent, but HbA1c was still 9.3% after 1 year of management. Her mother already had type 2 DM at 46-year-old and was on medication. Under the possibility of familial monogenic DM, targeted exome sequencing was performed which included 29 genes associated with monogenic DM. Nonsense mutation of the gene RFX6 (c.2661T>A, p.Tyr887∗) was found. After adding Glucagon-like peptide-1 (GLP-1) receptor agonist, HbA1c improved from 8.8 to 6.8% and body mass index (BMI) also improved from 31.0 to 29.2 kg/m2. CONCLUSIONS: It may be worth investigating genetic etiology in early-onset autoantibody-negative DM for specific genetic diagnosis and better management.


Sujet(s)
Codon non-sens , Diabète de type 2/anatomopathologie , Facteurs de transcription des facteurs régulateurs X/génétique , Adolescent , Diabète de type 2/traitement médicamenteux , Diabète de type 2/étiologie , Diabète de type 2/génétique , Femelle , Humains , Hypoglycémiants/usage thérapeutique , Adulte d'âge moyen , Pronostic
11.
J Diabetes Investig ; 12(10): 1914-1918, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-33721395

RÉSUMÉ

Heterozygous RFX6 mutation has emerged as a potential cause of maturity-onset diabetes mellitus of the young (MODY). A 16-year-old female was diagnosed with diabetes by her family doctor and was referred to our institution for genetic examination. Genetic testing revealed a novel RFX6 heterozygous mutation (NM_173560: exon17: c.1954C>T: p.R652X) in the patient and in her mother and brother. She had no islet-specific autoantibodies and showed a reduced meal-induced response of insulin, glucose-dependent insulinotropic polypeptide, and glucagon-like peptide-1, which is consistent with the phenotype of MODY due to heterozygous RFX6 mutation. In conclusion, we report a case of MODY due to a novel heterozygous mutation, p.R652X.


Sujet(s)
Diabète de type 2/génétique , Facteurs de transcription des facteurs régulateurs X/génétique , Adolescent , Diabète de type 2/sang , Femelle , Humains , Mutation , Pedigree
12.
J Mol Endocrinol ; 66(2): 129-140, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33350979

RÉSUMÉ

RFX6 transcription factor is believed to play a central role in directing cell development of insulin-producing pancreatic islet. RFX6 homozygous mutations cause syndromic neonatal diabetes with hypoplastic pancreas. However, RFX6 heterozygous mutations cause maturity-onset diabetes of the young (MODY) with normal pancreas development. Here, we show that RFX6 may control islet cell development and insulin production in different manners. The rfx6 knockout zebrafish generated by CRISPR/Cas9 exhibited an overt diabetes phenotype. Pancreatic islet failed to form compact structures in the knockout fish. While endocrine pancreatic islet non-ß-cells were absent, insulin-producing ß-cells were present in the knockout fish. Although insulin mRNA level was normal in the ß-cells of the knockout fish, insulin protein level was decreased. High-throughput RNA sequencing (RNAseq) showed that differentially expressed genes were enriched in the translation term in islet ß-cells from the knockout fish. Chromatin immunoprecipitation sequencing (ChIPseq) of normally developed islet ß-cells from mice demonstrated that rfx6 interacted with translation initiation factors and controlled insulin translation. Our data indicate that Rfx6 may act as a transcription factor regulating the transcription of genes involved in mRNA translation, which may represent a new mechanism and treatment strategy for diseases.


Sujet(s)
Cellules à insuline/métabolisme , Insuline/biosynthèse , Facteurs de transcription des facteurs régulateurs X/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/métabolisme , Animaux , Diabète expérimental/génétique , Régulation de l'expression des gènes , Techniques de knock-out de gènes , Gene Ontology , Insuline/génétique , Insuline/métabolisme , Souris , Mutation/génétique , Régions promotrices (génétique)/génétique , Liaison aux protéines , Biosynthèse des protéines/génétique , Facteurs de transcription des facteurs régulateurs X/génétique , Transcription génétique , Danio zébré/génétique , Protéines de poisson-zèbre/génétique
13.
Development ; 147(21)2020 11 05.
Article de Anglais | MEDLINE | ID: mdl-33033118

RÉSUMÉ

Mitchell-Riley syndrome (MRS) is caused by recessive mutations in the regulatory factor X6 gene (RFX6) and is characterised by pancreatic hypoplasia and neonatal diabetes. To determine why individuals with MRS specifically lack pancreatic endocrine cells, we micro-CT imaged a 12-week-old foetus homozygous for the nonsense mutation RFX6 c.1129C>T, which revealed loss of the pancreas body and tail. From this foetus, we derived iPSCs and show that differentiation of these cells in vitro proceeds normally until generation of pancreatic endoderm, which is significantly reduced. We additionally generated an RFX6HA reporter allele by gene targeting in wild-type H9 cells to precisely define RFX6 expression and in parallel performed in situ hybridisation for RFX6 in the dorsal pancreatic bud of a Carnegie stage 14 human embryo. Both in vitro and in vivo, we find that RFX6 specifically labels a subset of PDX1-expressing pancreatic endoderm. In summary, RFX6 is essential for efficient differentiation of pancreatic endoderm, and its absence in individuals with MRS specifically impairs formation of endocrine cells of the pancreas head and tail.


Sujet(s)
Différenciation cellulaire , Diabète/génétique , Diabète/anatomopathologie , Endoderme/embryologie , Maladies de la vésicule biliaire/génétique , Maladies de la vésicule biliaire/anatomopathologie , Cellules souches pluripotentes induites/anatomopathologie , Atrésie intestinale/génétique , Atrésie intestinale/anatomopathologie , Mutation/génétique , Pancréas/embryologie , Facteurs de transcription des facteurs régulateurs X/génétique , Allèles , Séquence nucléotidique , Différenciation cellulaire/génétique , Chromatine/métabolisme , Consanguinité , Diabète/imagerie diagnostique , Embryon de mammifère/métabolisme , Développement embryonnaire , Famille , Femelle , Maladies de la vésicule biliaire/imagerie diagnostique , Génome humain , Humains , Cellules souches pluripotentes induites/métabolisme , Atrésie intestinale/imagerie diagnostique , Mâle , Pedigree , Transcription génétique , Transcriptome/génétique , Microtomographie aux rayons X
14.
Diabetologia ; 63(10): 1974-1980, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32894307

RÉSUMÉ

Improving our understanding of mammalian pancreas development is crucial for the development of more effective cellular therapies for diabetes. Most of what we know about mammalian pancreas development stems from mouse genetics. We have learnt that a unique set of transcription factors controls endocrine and exocrine cell differentiation. Transgenic mouse models have been instrumental in studying the function of these transcription factors. Mouse and human pancreas development are very similar in many respects, but the devil is in the detail. To unravel human pancreas development in greater detail, in vitro cellular models (including directed differentiation of stem cells, human beta cell lines and human pancreatic organoids) are used; however, in vivo validation of these results is still needed. The current best 'model' for studying human pancreas development are individuals with monogenic forms of diabetes. In this review, we discuss mammalian pancreas development, highlight some discrepancies between mouse and human, and discuss selected transcription factors that, when mutated, cause permanent neonatal diabetes. Graphical abstract.


Sujet(s)
Régulation de l'expression des gènes au cours du développement/génétique , Pancréas/embryologie , Facteurs de transcription/génétique , Animaux , Lignée cellulaire , Diabète/génétique , Humains , Techniques in vitro , Cellules à insuline , Souris , Organoïdes , Pancréas/métabolisme , Cellules souches pluripotentes
15.
Mol Metab ; 29: 24-39, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31668390

RÉSUMÉ

OBJECTIVE: Enteroendocrine cells (EECs) of the gastro-intestinal tract sense gut luminal factors and release peptide hormones or serotonin (5-HT) to coordinate energy uptake and storage. Our goal is to decipher the gene regulatory networks controlling EECs specification from enteroendocrine progenitors. In this context, we studied the role of the transcription factor Rfx6 which had been identified as the cause of Mitchell-Riley syndrome, characterized by neonatal diabetes and congenital malabsorptive diarrhea. We previously reported that Rfx6 was essential for pancreatic beta cell development and function; however, the role of Rfx6 in EECs differentiation remained to be elucidated. METHODS: We examined the molecular, cellular, and metabolic consequences of constitutive and conditional deletion of Rfx6 in the embryonic and adult mouse intestine. We performed single cell and bulk RNA-Seq to characterize EECs diversity and identify Rfx6-regulated genes. RESULTS: Rfx6 is expressed in the gut endoderm; later, it is turned on in, and restricted to, enteroendocrine progenitors and persists in hormone-positive EECs. In the embryonic intestine, the constitutive lack of Rfx6 leads to gastric heterotopia, suggesting a role in the maintenance of intestinal identity. In the absence of intestinal Rfx6, EECs differentiation is severely impaired both in the embryo and adult. However, the number of serotonin-producing enterochromaffin cells and mucosal 5-HT content are increased. Concomitantly, Neurog3-positive enteroendocrine progenitors accumulate. Combined analysis of single-cell and bulk RNA-Seq data revealed that enteroendocrine progenitors differentiate in two main cell trajectories, the enterochromaffin (EC) cells and the Peptidergic Enteroendocrine (PE) cells, the differentiation programs of which are differentially regulated by Rfx6. Rfx6 operates upstream of Arx, Pax6 and Isl1 to trigger the differentiation of peptidergic EECs such as GIP-, GLP-1-, or CCK-secreting cells. On the contrary, Rfx6 represses Lmx1a and Tph1, two genes essential for serotonin biosynthesis. Finally, we identified transcriptional changes uncovering adaptive responses to the prolonged lack of enteroendocrine hormones and leading to malabsorption and lower food efficiency ratio in Rfx6-deficient mouse intestine. CONCLUSION: These studies identify Rfx6 as an essential transcriptional regulator of EECs specification and shed light on the molecular mechanisms of intestinal failures in human RFX6-deficiencies such as Mitchell-Riley syndrome.


Sujet(s)
Différenciation cellulaire , Facteurs de transcription des facteurs régulateurs X/métabolisme , Sérotonine/métabolisme , Animaux , Lignage cellulaire , Diarrhée/métabolisme , Diarrhée/anatomopathologie , Métabolisme énergétique , Cellules entérochromaffines/cytologie , Cellules entérochromaffines/métabolisme , Cellules entéroendocrines/cytologie , Cellules entéroendocrines/métabolisme , Femelle , Régulation de l'expression des gènes , Protéines à homéodomaine/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Protéines à homéodomaine LIM/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Facteurs de transcription des facteurs régulateurs X/déficit , Facteurs de transcription des facteurs régulateurs X/génétique , Analyse sur cellule unique , Facteurs de transcription/métabolisme
16.
Front Pediatr ; 7: 243, 2019.
Article de Anglais | MEDLINE | ID: mdl-31275908

RÉSUMÉ

We report a Saudi girl who presented at birth with neonatal diabetes, duodenal atresia, and progressive cholestasis. After other gene testing was negative, the clinical diagnosis of Mitchell-Riley syndrome was ultimately considered and further genetic analysis revealed a novel missense homozygous variant in RFX6: c.983A>T (p.asp328Val). Despite intensive management, the patient died from severe Klebsiella pneumoniae sepsis at 5 months of age. This rare syndrome should be suspected in any neonate with hyperglycemia complicated by intestinal atresia and/or progressive cholestasis that could suggest biliary hypoplasia. Early recognition and diagnosis through genetic testing are essential for guiding aggressive clinical management as well as family counseling, particularly in light of the high possibility of early death in this highly complex disorder.

17.
Biochem Biophys Res Commun ; 508(2): 556-562, 2019 01 08.
Article de Anglais | MEDLINE | ID: mdl-30509498

RÉSUMÉ

Regulatory Factor X-box binding transcriptional factor 6 (Rfx6) plays an important role in the differentiation and development of pancreas in mammals. However, the direct target genes of Rfx6 to regulate this process were largely unknown. The present study aimed to investigate the function of Rfx6 on regulating pancreatic differentiation and development in a physiologically-relevant context. We performed the chromatin immunoprecipitation followed by the next generation sequencing analysis (ChIP-seq) using whole pancreatic tissue harvested from C57/BL6 adult mice to find target genes of Rfx6. We captured 4146 unique peaks in the genome region of the adult murine pancreas. Among all these binding peaks, a majority were located in intron or intergenic regions. We further annotated all peaks to their nearest gene, and over 1000 genes were captured as Rfx6-binding genes in the pancreas. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis found that Rfx6-binding genes to be associated with the pancreas developmental process. A portion of selected ChIP-seq targets related with pancreas differentiation including Pdx1, Neurod1, Hnf1a, Nkx6-1, St18 and Shox2 were selected and validated as true targets by independent qPCR experiments. In addition, Rfx6 can directly bind to upstream of MiR-145, MiR-195, and possibly other non-protein-coding functional RNAs to control adult mouse pancreatic differentiation. Interestingly, our study revealed that Rfx6 played an important role in insulin translation by binding to the Eif2ak1, Upf1, and Eif5. Our data provide direct target genes of Rfx6 during pancreas development and point to Rfx6 as a potential therapeutic target for improving insulin protein content.


Sujet(s)
Séquençage nucléotidique à haut débit/méthodes , Insuline/biosynthèse , Pancréas/croissance et développement , Facteurs de transcription des facteurs régulateurs X/génétique , Animaux , Différenciation cellulaire , Régulation de l'expression des gènes , Insuline/métabolisme , Souris , Souris de lignée C57BL , Organogenèse , Pancréas/composition chimique , Liaison aux protéines , Facteurs de transcription des facteurs régulateurs X/métabolisme
18.
Diabetes Obes Metab ; 18 Suppl 1: 128-36, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27615142

RÉSUMÉ

Diabetes (either type 1 or type 2) is due to insufficient functional ß-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either ß-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model ß-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human ß-cells share many similarities with rodent ß-cells, they also differ on a number of important parameters. In this context, developing ways to study human ß-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human ß-cells that should be useful tools to discover new ways to modulate functional ß-cell mass in humans.


Sujet(s)
Techniques de culture cellulaire , Diabète/thérapie , Cellules souches embryonnaires humaines , Cellules à insuline/cytologie , Animaux , Recherche biomédicale , Différenciation cellulaire , Lignée cellulaire , Humains , Souris , Modèles animaux , Rats
19.
Eur J Med Genet ; 59(9): 429-35, 2016 Sep.
Article de Anglais | MEDLINE | ID: mdl-27523286

RÉSUMÉ

Mitchell-Riley syndrome, an autosomal recessive disorder caused by mutations in the RFX6 gene, is defined as a combination of neonatal diabetes mellitus and serious congenital gastrointestinal defects. We describe Mitchell-Riley syndrome in two sisters with two novel compound heterozygous variants in the RFX6 gene: c.1154G > A, p.(Arg385Gln), and c.1316_1319delTCTA, p.(Ile439Thrfs*13). Both sisters present milder forms of the syndrome, likely due to possible residual activity of the p.Arg385Gln variant, which is localized in a dimerization domain of the RFX6 transcription factor. We propose that the prognosis is dependent on patient RFX6 genotype and possible residual activity of RFX6 transcription factor. Both sisters had atypical later onset of diabetes, at 2 years and 10 months and 2 years and 7 months, respectively. This supports the need of extending the definition of diabetes in Mitchell-Riley syndrome from neonatal to childhood onset and regular glyceamia check in patients with gastrointestinal tract malformations typical for Mitchell-Riley syndrome. The clinical course in both sisters improved significantly after surgical removal of parts of the small intestine with heterotopic gastric mucosa. We suggest that gastric mucosa heterotopy is an important actionable part of Mitchell-Riley syndrome and could have been responsible for the malabsorption, failure to thrive and severe anemia present in previously reported patients with Mitchell-Riley syndrome.


Sujet(s)
Diabète/génétique , Maladies de la vésicule biliaire/génétique , Muqueuse gastrique/anatomopathologie , Atrésie intestinale/génétique , Facteurs de transcription des facteurs régulateurs X/génétique , Enfant , Enfant d'âge préscolaire , Diabète/étiologie , Diabète/anatomopathologie , Femelle , Maladies de la vésicule biliaire/étiologie , Maladies de la vésicule biliaire/anatomopathologie , Muqueuse gastrique/chirurgie , Hétérozygote , Humains , Atrésie intestinale/étiologie , Atrésie intestinale/anatomopathologie , Intestin grêle/malformations , Intestin grêle/chirurgie , Syndromes de malabsorption/génétique , Grossesse , Facteurs de transcription des facteurs régulateurs X/métabolisme , Fratrie
20.
Pediatr Diabetes ; 15(1): 67-72, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-23914949

RÉSUMÉ

Recently, bi-allelic mutations in the transcription factor RFX6 were described as the cause of a rare condition characterized by neonatal diabetes with pancreatic and biliary hypoplasia and duodenal/jejunal atresia. A male infant developed severe hyperglycemia (446 mg/dL) within 24 h of birth. Acute abdominal concerns by day five necessitated exploratory surgery that revealed duodenal atresia, gallbladder agenesis, annular pancreas and intestinal malrotation. He also exhibited chronic diarrhea and feeding intolerance, cholestatic jaundice, and subsequent liver failure. He died of sepsis at four months old while awaiting liver transplantation. The phenotype of neonatal diabetes with intestinal atresia and biliary agenesis clearly pointed to RFX6 as the causative gene; indeed, whole exome sequencing revealed a novel homozygous RFX6 mutation c.779A>C; p.Lys260Thr (K260T). This missense mutation also changes the consensus 5' splice donor site before intron 7 and is thus predicted to cause disruption in splicing. Both parents, who were not known to be related, were heterozygous carriers. Targeted genetic testing based on consideration of phenotypic features may reveal a cause among the many genes now associated with heterogeneous forms of monogenic neonatal diabetes. Our study demonstrates the feasibility of using modern sequencing technology to identify one such rare cause. Continued research is needed to determine the possible cost-effectiveness of this approach, especially when clear phenotypic clues are absent. Further study of patients with RFX6 mutations should clarify its role in pancreatic, intestinal and enteroendocrine cellular development and explain features such as the diarrhea exhibited in our case.


Sujet(s)
Malformations multiples/génétique , Protéines de liaison à l'ADN/génétique , Diabète de type 1/congénital , Maladies néonatales/génétique , Facteurs de transcription/génétique , Malformations , Malformations de l'appareil digestif , Occlusion duodénale , Issue fatale , Vésicule biliaire/malformations , Humains , Nourrisson , Nouveau-né , Atrésie intestinale , Volvulus intestinal , Mâle , Pancréas/malformations , Maladies du pancréas , Facteurs de transcription des facteurs régulateurs X
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE