Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Ann Hum Genet ; 84(3): 280-290, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31834638

RESUMO

Most genome-wide association studies used genetic-model-based tests assuming an additive mode of inheritance, leading to underpowered association tests in case of departure from additivity. The general regression model (GRM) association test proposed by Fisher and Wilson in 1980 makes no assumption on the genetic model. Interestingly, it also allows formal testing of the underlying genetic model. We conducted a simulation study of quantitative traits to compare the power of the GRM test to the classical linear regression tests, the maximum of the three statistics (MAX), and the allele-based (allelic) tests. Simulations were performed on two samples sizes, using a large panel of genetic models, varying genetic models, minor allele frequencies, and the percentage of explained variance. In case of departure from additivity, the GRM was more powerful than the additive regression tests (power gain reaching 80%) and had similar power when the true model is additive. GRM was also as or more powerful than the MAX or allelic tests. The true simulated model was mostly retained by the GRM test. Application of GRM to HbA1c illustrates its gain in power. To conclude, GRM increases power to detect association for quantitative traits, allows determining the genetic model and is easily applicable.


Assuntos
Estudo de Associação Genômica Ampla , Modelos Genéticos , Alelos , Simulação por Computador , Frequência do Gene , Hemoglobinas Glicadas/genética , Humanos , Modelos Lineares , Locos de Características Quantitativas
2.
Genomics ; 110(2): 98-111, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28911974

RESUMO

The GLIS family zinc finger 3 isoform (GLIS3) is a risk gene for Type 1 and Type 2 diabetes, glaucoma and Alzheimer's disease endophenotype. We identified GLIS3 binding sites in insulin secreting cells (INS1) (FDR q<0.05; enrichment range 1.40-9.11 fold) sharing the motif wrGTTCCCArTAGs, which were enriched in genes involved in neuronal function and autophagy and in risk genes for metabolic and neuro-behavioural diseases. We confirmed experimentally Glis3-mediated regulation of the expression of genes involved in autophagy and neuron function in INS1 and neuronal PC12 cells. Naturally-occurring coding polymorphisms in Glis3 in the Goto-Kakizaki rat model of type 2 diabetes were associated with increased insulin production in vitro and in vivo, suggestive alteration of autophagy in PC12 and INS1 and abnormal neurogenesis in hippocampus neurons. Our results support biological pleiotropy of GLIS3 in pathologies affecting ß-cells and neurons and underline the existence of trans­nosology pathways in diabetes and its co-morbidities.


Assuntos
Células Secretoras de Insulina/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Autofagia , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Hipocampo/citologia , Masculino , Neurogênese , Neurônios/citologia , Células PC12 , Polimorfismo Genético , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/química , Fatores de Transcrição/genética
3.
Diabetes Metab Res Rev ; 34(8): e3051, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30063815

RESUMO

Type 1 diabetes (T1D) results from autoimmune destruction of the pancreatic ßcells. Although all T1D patients require daily administration of exogenous insulin, their insulin requirement to achieve good glycaemic control may vary significantly. Glycated haemoglobin (HbA1c) level represents a stable indicator of glycaemic control and is a reliable predictor of long-term complications of T1D. The purpose of this article is to systematically review the role of non-genetic predictors and genetic factors of HbA1c level in T1D patients after the first year of T1D, to exclude the honeymoon period. A total of 1974 articles published since January 2011 were identified and 78 were finally included in the analysis of non-genetic predictors. For genetic factors, a total of 277 articles were identified and 14 were included. The most significantly associated factors with HbA1c level are demographic (age, ethnicity, and socioeconomic status), personal (family characteristics, parental care, psychological traits...) and features related to T1D (duration of T1D, adherence to treatment …). Only a few studies have searched for genetic factors influencing HbA1c level, most of which focused on candidate genes using classical genetic statistical methods, with generally limited power and incomplete adjustment for confounding factors and multiple testing. Our review shows the complexity of explaining HbA1c level variations, which involves numerous correlated predictors. Overall, our review underlines the lack of studies investigating jointly genetic and non-genetic factors and their interactions to better understand factors influencing glycaemic control for T1D patients.


Assuntos
Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Hemoglobinas Glicadas/genética , Hemoglobinas Glicadas/metabolismo , Adulto , Criança , Diabetes Mellitus Tipo 1/epidemiologia , Predisposição Genética para Doença , Humanos , Prognóstico , Fatores de Risco , Fatores de Tempo , Adulto Jovem
4.
PLoS Genet ; 9(5): e1003532, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23737756

RESUMO

Mutations in human Gli-similar (GLIS) 3 protein cause neonatal diabetes. The GLIS3 gene region has also been identified as a susceptibility risk locus for both type 1 and type 2 diabetes. GLIS3 plays a role in the generation of pancreatic beta cells and in insulin gene expression, but there is no information on the role of this gene on beta cell viability and/or susceptibility to immune- and metabolic-induced stress. GLIS3 knockdown (KD) in INS-1E cells, primary FACS-purified rat beta cells, and human islet cells decreased expression of MafA, Ins2, and Glut2 and inhibited glucose oxidation and insulin secretion, confirming the role of this transcription factor for the beta cell differentiated phenotype. GLIS3 KD increased beta cell apoptosis basally and sensitized the cells to death induced by pro-inflammatory cytokines (interleukin 1ß + interferon-γ) or palmitate, agents that may contribute to beta cell loss in respectively type 1 and 2 diabetes. The increased cell death was due to activation of the intrinsic (mitochondrial) pathway of apoptosis, as indicated by cytochrome c release to the cytosol, Bax translocation to the mitochondria and activation of caspases 9 and 3. Analysis of the pathways implicated in beta cell apoptosis following GLIS3 KD indicated modulation of alternative splicing of the pro-apoptotic BH3-only protein Bim, favouring expression of the pro-death variant BimS via inhibition of the splicing factor SRp55. KD of Bim abrogated the pro-apoptotic effect of GLIS3 loss of function alone or in combination with cytokines or palmitate. The present data suggest that altered expression of the candidate gene GLIS3 may contribute to both type 1 and 2 type diabetes by favouring beta cell apoptosis. This is mediated by alternative splicing of the pro-apoptotic protein Bim and exacerbated formation of the most pro-apoptotic variant BimS.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Idoso , Processamento Alternativo/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteínas de Ligação a DNA , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 2/etiologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Proteínas Repressoras , Transativadores
5.
PLoS Genet ; 9(10): e1003888, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204302

RESUMO

We describe a new syndrome of young onset diabetes, short stature and microcephaly with intellectual disability in a large consanguineous family with three affected children. Linkage analysis and whole exome sequencing were used to identify the causal nonsense mutation, which changed an arginine codon into a stop at position 127 of the tRNA methyltransferase homolog gene TRMT10A (also called RG9MTD2). TRMT10A mRNA and protein were absent in lymphoblasts from the affected siblings. TRMT10A is ubiquitously expressed but enriched in brain and pancreatic islets, consistent with the tissues affected in this syndrome. In situ hybridization studies showed that TRMT10A is expressed in human embryonic and fetal brain. TRMT10A is the mammalian ortholog of S. cerevisiae TRM10, previously shown to catalyze the methylation of guanine 9 (m(1)G9) in several tRNAs. Consistent with this putative function, in silico topology prediction indicated that TRMT10A has predominant nuclear localization, which we experimentally confirmed by immunofluorescence and confocal microscopy. TRMT10A localizes to the nucleolus of ß- and non-ß-cells, where tRNA modifications occur. TRMT10A silencing induces rat and human ß-cell apoptosis. Taken together, we propose that TRMT10A deficiency negatively affects ß-cell mass and the pool of neurons in the developing brain. This is the first study describing the impact of TRMT10A deficiency in mammals, highlighting a role in the pathogenesis of microcephaly and early onset diabetes. In light of the recent report that the type 2 diabetes candidate gene CDKAL1 is a tRNA methylthiotransferase, the findings in this family suggest broader relevance of tRNA methyltransferases in the pathogenesis of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Deficiência Intelectual/genética , Metiltransferases/genética , Microcefalia/genética , tRNA Metiltransferases/genética , Adulto , Idade de Início , Animais , Apoptose/genética , Diabetes Mellitus Tipo 2/complicações , Feminino , Ligação Genética , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Deficiência Intelectual/complicações , Deficiência Intelectual/patologia , Masculino , Microcefalia/complicações , Microcefalia/patologia , Mutação , Linhagem , Ratos , Proteínas de Saccharomyces cerevisiae/genética , tRNA Metiltransferases/deficiência
6.
Nat Genet ; 38(6): 682-7, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16715098

RESUMO

We recently described a new neonatal diabetes syndrome associated with congenital hypothyroidism, congenital glaucoma, hepatic fibrosis and polycystic kidneys. Here, we show that this syndrome results from mutations in GLIS3, encoding GLI similar 3, a recently identified transcription factor. In the original family, we identified a frameshift mutation predicted to result in a truncated protein. In two other families with an incomplete syndrome, we found that affected individuals harbor deletions affecting the 11 or 12 5'-most exons of the gene. The absence of a major transcript in the pancreas and thyroid (deletions from both families) and an eye-specific transcript (deletion from one family), together with residual expression of some GLIS3 transcripts, seems to explain the incomplete clinical manifestations in these individuals. GLIS3 is expressed in the pancreas from early developmental stages, with greater expression in beta cells than in other pancreatic tissues. These results demonstrate a major role for GLIS3 in the development of pancreatic beta cells and the thyroid, eye, liver and kidney.


Assuntos
Hipotireoidismo Congênito/genética , Diabetes Mellitus/genética , Doenças do Recém-Nascido/genética , Mutação , Fatores de Transcrição/genética , Alelos , Animais , Proteínas de Ligação a DNA , Feminino , Humanos , Recém-Nascido , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome , Transativadores
7.
Nat Genet ; 37(5): 507-13, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15838506

RESUMO

Dengue fever and dengue hemorrhagic fever are mosquito-borne viral diseases. Dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN1, encoded by CD209), an attachment receptor of dengue virus, is essential for productive infection of dendritic cells. Here, we report strong association between a promoter variant of CD209, DCSIGN1-336, and risk of dengue fever compared with dengue hemorrhagic fever or population controls. The G allele of the variant DCSIGN1-336 was associated with strong protection against dengue fever in three independent cohorts from Thailand, with a carrier frequency of 4.7% in individuals with dengue fever compared with 22.4% in individuals with dengue hemorrhagic fever (odds ratio for risk of dengue hemorrhagic fever versus dengue fever: 5.84, P = 1.4 x 10(-7)) and 19.5% in controls (odds ratio for protection: 4.90, P = 2 x 10(-6)). This variant affects an Sp1-like binding site and transcriptional activity in vitro. These results indicate that CD209 has a crucial role in dengue pathogenesis, which discriminates between severe dengue fever and dengue hemorrhagic fever. This may have consequences for therapeutic and preventive strategies.


Assuntos
Moléculas de Adesão Celular/genética , Dengue/genética , Lectinas Tipo C/genética , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Índice de Gravidade de Doença , Dengue/fisiopatologia , Humanos , Polimorfismo Genético
8.
Cells ; 11(3)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35159392

RESUMO

Human pluripotent stem cells, with their ability to proliferate indefinitely and to differentiate into virtually all cell types of the human body, provide a novel resource to study human development and to implement relevant disease models. Here, we employed a human pancreatic differentiation platform complemented with an shRNA screen in human pluripotent stem cells (PSCs) to identify potential drivers of early endoderm and pancreatic development. Deep sequencing followed by abundancy ranking pinpointed six top hit genes potentially associated with either improved or impaired endodermal differentiation, which were selected for functional validation in CRISPR-Cas9 mediated knockout (KO) lines. Upon endoderm differentiation (DE), particularly the loss of SLC22A1 and DSC2 led to impaired differentiation efficiency into CXCR4/KIT-positive DE cells. qPCR analysis also revealed changes in differentiation markers CXCR4, FOXA2, SOX17, and GATA6. Further differentiation of PSCs to the pancreatic progenitor (PP) stage resulted in a decreased proportion of PDX1/NKX6-1-positive cells in SLC22A1 KO lines, and in DSC2 KO lines when differentiated under specific culture conditions. Taken together, our study reveals novel genes with potential roles in early endodermal development.


Assuntos
Endoderma , Células-Tronco Pluripotentes , Diferenciação Celular/genética , Genômica , Humanos , Pâncreas/metabolismo , Células-Tronco Pluripotentes/metabolismo
9.
Front Endocrinol (Lausanne) ; 12: 648284, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34079523

RESUMO

Diabetes, as one of the major diseases in industrial countries, affects over 350 million people worldwide. Type 1 (T1D) and type 2 diabetes (T2D) are the most common forms with both types having invariable genetic influence. It is accepted that a subset of all diabetes patients, generally estimated to account for 1-2% of all diabetic cases, is attributed to mutations in single genes. As only a subset of these genes has been identified and fully characterized, there is a dramatic need to understand the pathophysiological impact of genetic determinants on ß-cell function and pancreatic development but also on cell replacement therapies. Pluripotent stem cells differentiated along the pancreatic lineage provide a valuable research platform to study such genes. This review summarizes current perspectives in applying this platform to study monogenic diabetes variants.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Mutação , Células-Tronco Pluripotentes/citologia , Animais , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Epigênese Genética , Edição de Genes , Variação Genética , Heterozigoto , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Pâncreas/embriologia , Pâncreas/patologia , Fenótipo , Regeneração
10.
Commun Biol ; 4(1): 1298, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789845

RESUMO

Cell type specification during pancreatic development is tightly controlled by a transcriptional and epigenetic network. The precise role of most transcription factors, however, has been only described in mice. To convey such concepts to human pancreatic development, alternative model systems such as pancreatic in vitro differentiation of human pluripotent stem cells can be employed. Here, we analyzed stage-specific RNA-, ChIP-, and ATAC-sequencing data to dissect transcriptional and regulatory mechanisms during pancreatic development. Transcriptome and open chromatin maps of pancreatic differentiation from human pluripotent stem cells provide a stage-specific pattern of known pancreatic transcription factors and indicate ONECUT1 as a crucial fate regulator in pancreas progenitors. Moreover, our data suggest that ONECUT1 is also involved in preparing pancreatic progenitors for later endocrine specification. The dissection of the transcriptional and regulatory circuitry revealed an important role for ONECUT1 within such network and will serve as resource to study human development and disease.


Assuntos
Fator 6 Nuclear de Hepatócito/genética , Pâncreas/fisiologia , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Fator 6 Nuclear de Hepatócito/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Transcrição Gênica
11.
J Neurol ; 268(7): 2515-2522, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33544221

RESUMO

The common co-occurrence of autoimmune systemic diseases in patients with neurological disorders and antibodies against glutamic acid decarboxylase 65 (GAD65) suggests a shared genetic predisposition to these disorders. However, the nature and frequency of familial aggregation of autoimmune diseases, which might also support this hypothesis, have been poorly investigated. Herein, an exploratory, interview-based study was conducted with the aim of describing the autoimmune diseases displayed by the relatives of GAD65 neurological patients, their frequency, kinship, and potential patterns of inheritance. Patients were enrolled only if they had GAD65 antibodies in the cerebrospinal fluid and typical clinical phenotypes associated with such antibodies (stiff-person syndrome, cerebellar ataxia, limbic encephalitis, or temporal lobe epilepsy). A total of 65 patients were included in the study, and 44/65 (67.7%) reported family history of autoimmunity, including first-degree relatives in 36/65 (55.4%); the sibling recurrence risk (λS) was 5.5, reinforcing the hypothesis of an underlying strong genetic predisposition. Most pedigrees with familial autoimmunity (38/44, 86.4%) showed multiple autoimmune diseases, all but 2 of them with diabetes mellitus or autoimmune thyroid disease, therefore resembling autoimmune polyendocrine syndromes. Inheritance patterns were diverse, possibly autosomal dominant in 17/44 (38.6%) pedigrees or autosomal recessive in 5/44 (11.4%), and un-defined or complex in 24/44 (54.5%). However, a total of 21/65 (32.3%) patients had no identified family history of autoimmunity. In conclusion, these results suggest a variable and heterogeneous genetic predisposition to GAD65 neurological disorders, possibly involving multiple loci and modes of inheritance with different contribution in each family.


Assuntos
Doenças Autoimunes , Rigidez Muscular Espasmódica , Autoanticorpos , Autoimunidade/genética , Glutamato Descarboxilase , Humanos
12.
Eur J Endocrinol ; 184(3): 455-468, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33486469

RESUMO

OBJECTIVE: DNAJC3, also known as P58IPK, is an Hsp40 family member that interacts with and inhibits PKR-like ER-localized eIF2α kinase (PERK). Dnajc3 deficiency in mice causes pancreatic ß-cell loss and diabetes. Loss-of-function mutations in DNAJC3 cause early-onset diabetes and multisystemic neurodegeneration. The aim of our study was to investigate the genetic cause of early-onset syndromic diabetes in two unrelated patients, and elucidate the mechanisms of ß-cell failure in this syndrome. METHODS: Whole exome sequencing was performed and identified variants were confirmed by Sanger sequencing. DNAJC3 was silenced by RNAi in INS-1E cells, primary rat ß-cells, human islets, and induced pluripotent stem cell-derived ß-cells. ß-cell function and apoptosis were assessed, and potential mediators of apoptosis examined. RESULTS: The two patients presented with juvenile-onset diabetes, short stature, hypothyroidism, neurodegeneration, facial dysmorphism, hypoacusis, microcephaly and skeletal bone deformities. They were heterozygous compound and homozygous for novel loss-of-function mutations in DNAJC3. DNAJC3 silencing did not impair insulin content or secretion. Instead, the knockdown induced rat and human ß-cell apoptosis and further sensitized cells to endoplasmic reticulum stress, triggering mitochondrial apoptosis via the pro-apoptototic Bcl-2 proteins BIM and PUMA. CONCLUSIONS: This report confirms previously described features and expands the clinical spectrum of syndromic DNAJC3 diabetes, one of the five monogenic forms of diabetes pertaining to the PERK pathway of the endoplasmic reticulum stress response. DNAJC3 deficiency may lead to ß-cell loss through BIM- and PUMA-dependent activation of the mitochondrial pathway of apoptosis.


Assuntos
Apoptose/genética , Diabetes Mellitus Tipo 1/genética , Proteínas de Choque Térmico HSP40/genética , Células Secretoras de Insulina/fisiologia , Mitocôndrias/metabolismo , Adolescente , Adulto , Fatores Etários , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/metabolismo , Mutação com Perda de Função , Masculino , Camundongos , Mitocôndrias/patologia , Linhagem , Ratos , Síndrome
13.
Nat Med ; 27(11): 1928-1940, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34663987

RESUMO

Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Fator 6 Nuclear de Hepatócito/genética , Pâncreas/embriologia , Diferenciação Celular/genética , Anormalidades Congênitas/genética , Retardo do Crescimento Fetal/genética , Vesícula Biliar/anormalidades , Proteína Homeobox Nkx-2.2/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Lactente , Recém-Nascido , Masculino , Herança Multifatorial/genética , Organogênese/genética , Pâncreas/anormalidades , Pancreatopatias/congênito , Pancreatopatias/genética , Células-Tronco Pluripotentes/citologia , Transcrição Gênica/genética
14.
Hum Mol Genet ; 17(24): 4012-21, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18806274

RESUMO

Most cases of juvenile-onset diabetes (JOD) are diagnosed as type 1 diabetes (T1D), for which genetic studies conducted in outbred Caucasian populations support the concept of multifactorial inheritance. However, this view may be partly challenged in particular population settings. In view of the suggestive evidence for a high prevalence of Wolfram syndrome (WFS) in Lebanon, the phenotypic variability associated with WFS1 mutations, and the high consanguinity rate in Lebanon, we aimed to evaluate the contribution of WFS1 mutations as monogenic determinants to JOD in Lebanon. We performed a family-based genetic study, with linkage analysis followed by systematic mutation screening of WFS1 exons in all JOD probands. The study population consisted of an unbiased recruitment of all juvenile-onset insulin-dependent diabetic patients from a specialized diabetes pediatric clinic in Beirut, Lebanon. Homozygous or compound heterozygous WFS1 mutations were found in 22 of the 399 JOD probands (5.5%), resulting in WFS (17 probands) or in non-syndromic non-autoimmune diabetes mellitus (DM, five probands). These accounted for 12.1% (21/174) of probands in consanguineous families, compared with 0.4% (1/225) in non-consanguineous families. Of the 38 patients identified with homozygous or compound heterozygous WFS1 mutations, 11 (29%) had non-syndromic DM, all of whom carried a particular WFS1 mutation, WFS1(LIB), encoding a protein with an extended C-terminal domain. This mutation resulted in a delayed onset or absence of extrapancreatic features. These results underscore the major impact of population-specific factors, such as population-specific mutations and founder effects, and family structure in the genetic determinism of JOD.


Assuntos
Diabetes Mellitus Tipo 1/genética , Proteínas de Membrana/genética , Mutação/genética , Adolescente , Adulto , Autoanticorpos/biossíntese , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/epidemiologia , Éxons/genética , Ligação Genética , Marcadores Genéticos/genética , Predisposição Genética para Doença , Variação Genética , Humanos , Lactente , Líbano/epidemiologia , Prevalência , Síndrome , Síndrome de Wolfram/epidemiologia , Síndrome de Wolfram/genética , Adulto Jovem
15.
Pediatr Diabetes ; 11(4): 279-85, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20202148

RESUMO

Wolcott-Rallison syndrome (WRS) is a rare autosomal recessive disorder characterized by an early-infancy-onset diabetes mellitus associated with a variety of multisystemic clinical manifestations. Here, we present six patients with WRS, carrying the same homozygous mutation (EIF2AK3-W522X), from two unrelated Turkish families. This is the largest series of patients with the same mutation for this rare syndrome. In this communication we compare clinical features of these six patients with the other 34 patients who have been reported to date, and review the clinical features of WRS. All WRS patients presented first with symptoms of insulin dependent diabetes mellitus, with a mean age at onset of 2 months. All patients had skeletal dysplasia or early signs of it, and growth retardation. Many of the patients with WRS have been reported to have developmental delay, mental retardation, and learning difficulties; in contrast, none of our patients showed abnormal development at age up to 30 months. Acute attacks of hepatic failure were reported in 23 cases out of 37 patients; in 15 of those 23 cases an acute attack of renal failure accompanied the liver failure. Exocrine pancreatic deficiency has been reported in only four cases other than our four patients. Central hypothyroidism was observed in six of 28 cases. We propose that central hypothyroidism is not a component of WRS, but rather a reflection of euthyroid sick syndrome. Four of our patients experienced severe neutropenia, compared to only five of the 27 other cases, suggesting that the W522X mutation may be specifically associated with neutropenia. Other than the consistent features of diabetes mellitus and epiphyseal dysplasia, WRS patients are otherwise characterized by extensive phenotypic variability that correlates poorly to genotype.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Mutação Puntual , eIF-2 Quinase/genética , Epífises/anormalidades , Evolução Fatal , Feminino , Humanos , Hipotireoidismo/diagnóstico , Hipotireoidismo/genética , Lactente , Falência Hepática/genética , Masculino , Neutropenia/diagnóstico , Neutropenia/genética , Pâncreas Exócrino/enzimologia , Linhagem
16.
J Clin Invest ; 130(12): 6338-6353, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33164986

RESUMO

Neonatal diabetes is caused by single gene mutations reducing pancreatic ß cell number or impairing ß cell function. Understanding the genetic basis of rare diabetes subtypes highlights fundamental biological processes in ß cells. We identified 6 patients from 5 families with homozygous mutations in the YIPF5 gene, which is involved in trafficking between the endoplasmic reticulum (ER) and the Golgi. All patients had neonatal/early-onset diabetes, severe microcephaly, and epilepsy. YIPF5 is expressed during human brain development, in adult brain and pancreatic islets. We used 3 human ß cell models (YIPF5 silencing in EndoC-ßH1 cells, YIPF5 knockout and mutation knockin in embryonic stem cells, and patient-derived induced pluripotent stem cells) to investigate the mechanism through which YIPF5 loss of function affects ß cells. Loss of YIPF5 function in stem cell-derived islet cells resulted in proinsulin retention in the ER, marked ER stress, and ß cell failure. Partial YIPF5 silencing in EndoC-ßH1 cells and a patient mutation in stem cells increased the ß cell sensitivity to ER stress-induced apoptosis. We report recessive YIPF5 mutations as the genetic cause of a congenital syndrome of microcephaly, epilepsy, and neonatal/early-onset diabetes, highlighting a critical role of YIPF5 in ß cells and neurons. We believe this is the first report of mutations disrupting the ER-to-Golgi trafficking, resulting in diabetes.


Assuntos
Diabetes Mellitus , Estresse do Retículo Endoplasmático/genética , Doenças Genéticas Inatas , Doenças do Recém-Nascido , Microcefalia , Mutação , Proteínas de Transporte Vesicular , Linhagem Celular , Diabetes Mellitus/embriologia , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Feminino , Doenças Genéticas Inatas/embriologia , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Recém-Nascido , Doenças do Recém-Nascido/embriologia , Doenças do Recém-Nascido/genética , Doenças do Recém-Nascido/patologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Microcefalia/embriologia , Microcefalia/genética , Microcefalia/patologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
17.
Diabetes ; 56(2): 522-6, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17259401

RESUMO

The PTPN22 gene, encoding the lymphoid-specific protein tyrosine phosphatase, a negative regulator in the T-cell activation and development, has been associated with the susceptibility to several autoimmune diseases, including type 1 diabetes. Based on combined case-control and family-based association studies, we replicated the finding of an association of the PTPN22 C1858T (R620W) functional variant with type 1 diabetes, which was independent from the susceptibility status at the insulin gene and at HLA-DR (DR3/4 compared with others). The risk contributed by the 1858T allele was increased in patients with a family history of other autoimmune diseases, further supporting a general role for this variant on autoimmunity. In addition, we found evidence for an association of 1858T allele with the presence of GAD autoantibodies (GADA), which was restricted to patients with long disease duration (>10 years, P < 0.001). This may help define a subgroup of patients with long-term persistence of GADA. The risk conferred by 1858T allele on GAD positivity was additive, and our meta-analysis also supported an additive rather than dominant effect of this variant on type 1 diabetes, similar to previous reports on rheumatoid arthritis and systemic lupus erythematosus.


Assuntos
Autoimunidade/genética , Proteínas de Ligação a DNA/imunologia , Diabetes Mellitus Tipo 1/genética , Predisposição Genética para Doença , Glutamato Descarboxilase/imunologia , Fragmentos de Peptídeos/imunologia , Proteínas Tirosina Fosfatases/genética , Proteínas Repressoras/imunologia , Alelos , Autoanticorpos/genética , Biomarcadores , Estudos de Casos e Controles , Predisposição Genética para Doença/genética , Genótipo , Antígenos HLA-DR/genética , Humanos , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteína Tirosina Fosfatase não Receptora Tipo 22
18.
Biomed Pharmacother ; 62(3): 153-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17961971

RESUMO

A functional variant located in the promoter region of the Fc receptor like 3 gene (FCRL3, -169CT variant) has been recently shown to be associated with several autoimmune diseases in the Japanese population. Following the concept of shared genetic determinants between autoimmune diseases, we tested this variant for association to Type 1 diabetes (T1D) and T1D-related phenotypes in two independent settings: a family-based association study (French and US families) and a case-control study (French population). We found suggestive evidence for association of the FCRL3 -169CC genotype, corresponding to the susceptibility genotype for rheumatoid arthritis, with an increased risk of additional autoimmunity markers (OR=1.97, P=0.01) and of other autoimmune diseases (OR=1.75, P=0.05). However, there was no evidence of association of this variant with T1D in these cohorts, separately and combined, nor in subgroups of patients defined based on their major T1D risk factors at HLA-DRB1, insulin and PTPN22. Hence, this variant may help define subgroups of T1D patients with contrasted risk of other autoimmune diseases.


Assuntos
Doenças Autoimunes/genética , Diabetes Mellitus Tipo 1/genética , Receptores Imunológicos/genética , Doenças Autoimunes/epidemiologia , Estudos de Casos e Controles , Estudos de Coortes , Diabetes Mellitus Tipo 1/epidemiologia , França/epidemiologia , Genótipo , Haplótipos , Humanos , Polimorfismo Genético , Fatores de Risco , Estados Unidos/epidemiologia
19.
Genes (Basel) ; 8(11)2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29112131

RESUMO

Monogenic forms of diabetes may account for 1-5% of all cases of diabetes, and may occur in the context of syndromic presentations. We investigated the case of a girl affected by insulin-dependent diabetes, diagnosed at 6 years old, associated with congenital cataract. Her consanguineous parents and her four other siblings did not have diabetes or cataract, suggesting a recessive syndrome. Using whole exome sequencing of the affected proband, we identified a heterozygous p.R825Q ABCC8 mutation, located at the exact same amino-acid position as the p.R825W recurring diabetes mutation, hence likely responsible for the diabetes condition, and a homozygous p.G71S mutation in CRYBB1, a gene known to be responsible for congenital cataract. Both mutations were predicted to be damaging and were absent or extremely rare in public databases. Unexpectedly, we found that the mother was also homozygous for the CRYBB1 mutation, and both the mother and one unaffected sibling were heterozygous for the ABCC8 mutation, suggesting incomplete penetrance of both mutations. Incomplete penetrance of ABCC8 mutations is well documented, but this is the first report of an incomplete penetrance of a CRYBB1 mutation, manifesting between susceptible subjects (unaffected mother vs. affected child) and to some extent within the patient herself, who had distinct cataract severities in both eyes. Our finding illustrates the importance of family studies to unmask the role of confounding factors such as double-gene mutations and incomplete penetrance that may mimic monogenic syndromes including in the case of strongly evocative family structure with consanguinity.

20.
Diabetes ; 66(4): 1086-1096, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28073829

RESUMO

We describe a new syndrome characterized by early-onset diabetes associated with bone marrow failure, affecting mostly the erythrocytic lineage. Using whole-exome sequencing in a remotely consanguineous patient from a family with two affected siblings, we identified a single homozygous missense mutation (chr15.hg19:g.48,626,619A>G) located in the dUTPase (DUT) gene (National Center for Biotechnology Information Gene ID 1854), affecting both the mitochondrial (DUT-M p.Y142C) and the nuclear (DUT-N p.Y54C) isoforms. We found the same homozygous mutation in an unrelated consanguineous patient with diabetes and bone marrow aplasia from a family with two affected siblings, whereas none of the >60,000 subjects from the Exome Aggregation Consortium (ExAC) was homozygous for this mutation. This replicated observation probability was highly significant, thus confirming the role of this DUT mutation in this syndrome. DUT is a key enzyme for maintaining DNA integrity by preventing misincorporation of uracil into DNA, which results in DNA toxicity and cell death. We showed that DUT silencing in human and rat pancreatic ß-cells results in apoptosis via the intrinsic cell death pathway. Our findings support the importance of tight control of DNA metabolism for ß-cell integrity and warrant close metabolic monitoring of patients treated by drugs affecting dUTP balance.


Assuntos
Anemia Aplástica/genética , Apoptose/genética , Doenças da Medula Óssea/genética , Diabetes Mellitus/genética , Hemoglobinúria Paroxística/genética , Pirofosfatases/genética , RNA Mensageiro/metabolismo , Adolescente , Adulto , Idoso , Animais , Western Blotting , Transtornos da Insuficiência da Medula Óssea , Criança , Consanguinidade , Cristalografia por Raios X , Feminino , Humanos , Ilhotas Pancreáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Estrutura Molecular , Mutação , RNA Interferente Pequeno , Ratos , Ratos Wistar , Análise de Sequência de DNA , Síndrome , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa