Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Med Genet ; 60(4): 337-345, 2023 04.
Article in English | MEDLINE | ID: mdl-35927022

ABSTRACT

BACKGROUND: Ellis-Van Creveld (EVC) syndrome is one of the entities belonging to the skeletal ciliopathies short rib-polydactyly subgroup. Major signs are ectodermal dysplasia, chondrodysplasia, polydactyly and congenital cardiopathy, with a high degree of variability in phenotypes ranging from lethal to mild clinical presentations. The EVC and EVC2 genes are the major genes causative of EVC syndrome. However, an increased number of genes involved in the ciliopathy complex have been identified in EVC syndrome, leading to a better understanding of its physiopathology, namely, WDR35, GLI1, DYNC2LI1, PRKACA, PRKACB and SMO. They all code for proteins located in the primary cilia, playing a key role in signal transduction of the Hedgehog pathways. METHODS: The aim of this study was the analysis of 50 clinically identified EVC cases from 45 families to further define the phenotype and molecular bases of EVC. RESULTS: Our detection rate in the cohort of 45 families was of 91.11%, with variants identified in EVC/EVC2 (77.8%), DYNC2H1 (6.7%), DYNC2LI1 (2.2%), SMO (2.2%) or PRKACB (2.2%). No distinctive feature was remarkable of a specific genotype-phenotype correlation. Interestingly, we identified a high proportion of heterozygous deletions in EVC/EVC2 of variable sizes (26.92%), mostly inherited from the mother, and probably resulting from recombinations involving Alu sequences. CONCLUSION: We confirmed that EVC and EVC2 are the major genes involved in the EVC phenotype and highlighted the high prevalence of previously unreported CNVs (Copy Number Variation).


Subject(s)
Ellis-Van Creveld Syndrome , Polydactyly , Humans , Hedgehog Proteins/genetics , Ellis-Van Creveld Syndrome/genetics , Ellis-Van Creveld Syndrome/diagnosis , DNA Copy Number Variations/genetics , Phenotype
2.
Gut ; 71(3): 497-508, 2022 03.
Article in English | MEDLINE | ID: mdl-33579790

ABSTRACT

OBJECTIVE: Enteropathy-associated T-cell lymphoma (EATL) is a rare but severe complication of coeliac disease (CeD), often preceded by low-grade clonal intraepithelial lymphoproliferation, referred to as type II refractory CeD (RCDII). Knowledge on underlying oncogenic mechanisms remains scarce. Here, we analysed and compared the mutational landscape of RCDII and EATL in order to identify genetic drivers of CeD-associated lymphomagenesis. DESIGN: Pure populations of RCDII-cells derived from intestinal biopsies (n=9) or sorted from blood (n=2) were analysed by whole exome sequencing, comparative genomic hybridisation and RNA sequencing. Biopsies from RCDII (n=50), EATL (n=19), type I refractory CeD (n=7) and uncomplicated CeD (n=18) were analysed by targeted next-generation sequencing. Moreover, functional in vitro studies and drug testing were performed in RCDII-derived cell lines. RESULTS: 80% of RCDII and 90% of EATL displayed somatic gain-of-functions mutations in the JAK1-STAT3 pathway, including a remarkable p.G1097 hotspot mutation in the JAK1 kinase domain in approximately 50% of cases. Other recurrent somatic events were deleterious mutations in nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) regulators TNFAIP3 and TNIP3 and potentially oncogenic mutations in TET2, KMT2D and DDX3X. JAK1 inhibitors, and the proteasome inhibitor bortezomib could block survival and proliferation of malignant RCDII-cell lines. CONCLUSION: Mutations activating the JAK1-STAT3 pathway appear to be the main drivers of CeD-associated lymphomagenesis. In concert with mutations in negative regulators of NF-κB, they may favour the clonal emergence of malignant lymphocytes in the cytokine-rich coeliac intestine. The identified mutations are attractive therapeutic targets to treat RCDII and block progression towards EATL.


Subject(s)
Celiac Disease/complications , Celiac Disease/genetics , Enteropathy-Associated T-Cell Lymphoma/etiology , Gain of Function Mutation/genetics , Lymphocytes/physiology , Adult , Aged , Aged, 80 and over , Celiac Disease/pathology , Cohort Studies , Enteropathy-Associated T-Cell Lymphoma/pathology , Female , France , Humans , Janus Kinase 1/genetics , Male , Middle Aged , STAT3 Transcription Factor/genetics , Young Adult
3.
Am J Hum Genet ; 101(6): 1006-1012, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29198720

ABSTRACT

Leber congenital amaurosis (LCA) is a neurodegenerative disease of photoreceptor cells that causes blindness within the first year of life. It occasionally occurs in syndromic metabolic diseases and plurisystemic ciliopathies. Using exome sequencing in a multiplex family and three simplex case subjects with an atypical association of LCA with early-onset hearing loss, we identified two heterozygous mutations affecting Arg391 in ß-tubulin 4B isotype-encoding (TUBB4B). Inspection of the atomic structure of the microtubule (MT) protofilament reveals that the ß-tubulin Arg391 residue contributes to a binding pocket that interacts with α-tubulin contained in the longitudinally adjacent αß-heterodimer, consistent with a role in maintaining MT stability. Functional analysis in cultured cells overexpressing FLAG-tagged wild-type or mutant TUBB4B as well as in primary skin-derived fibroblasts showed that the mutant TUBB4B is able to fold, form αß-heterodimers, and co-assemble into the endogenous MT lattice. However, the dynamics of growing MTs were consistently altered, showing that the mutations have a significant dampening impact on normal MT growth. Our findings provide a link between sensorineural disease and anomalies in MT behavior and describe a syndromic LCA unrelated to ciliary dysfunction.


Subject(s)
Leber Congenital Amaurosis/genetics , Microtubules/genetics , Tubulin/genetics , Adult , Binding Sites/genetics , Cells, Cultured , Child , DNA Mutational Analysis , Female , Humans , Male , Microtubules/metabolism , Middle Aged , Mutation, Missense/genetics , Photoreceptor Cells/metabolism , Tubulin/metabolism , Exome Sequencing
4.
Am J Med Genet A ; 176(1): 181-186, 2018 01.
Article in English | MEDLINE | ID: mdl-29159987

ABSTRACT

We report two unrelated patients with Pierre Robin sequence (PRS) and a strikingly similar combination of associated features. Whole exome sequencing was performed for both patients. No single gene containing likely pathogenic point mutations in both patients could be identified, but the finding of an essential splice site mutation in mediator complex subunit 13 like (MED13L) in one patient prompted the investigation of copy number variants in MED13L in the other, leading to the identification of an intragenic deletion. Disruption of MED13L, encoding a component of the Mediator complex, is increasingly recognized as the cause of an intellectual disability syndrome with associated facial dysmorphism. Our findings suggest that MED13L-related disorders are a possible differential diagnosis for syndromic PRS.


Subject(s)
Loss of Function Mutation , Mediator Complex/genetics , Pierre Robin Syndrome/diagnosis , Pierre Robin Syndrome/genetics , Brain/abnormalities , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Infant , Magnetic Resonance Imaging/methods , Male , Phenotype , Sequence Analysis, DNA
5.
J Am Soc Nephrol ; 28(10): 2901-2914, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28566479

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) occur in three to six of 1000 live births, represent about 20% of the prenatally detected anomalies, and constitute the main cause of CKD in children. These disorders are phenotypically and genetically heterogeneous. Monogenic causes of CAKUT in humans and mice have been identified. However, despite high-throughput sequencing studies, the cause of the disease remains unknown in most patients, and several studies support more complex inheritance and the role of environmental factors and/or epigenetics in the pathophysiology of CAKUT. Here, we report the targeted exome sequencing of 330 genes, including genes known to be involved in CAKUT and candidate genes, in a cohort of 204 unrelated patients with CAKUT; 45% of the patients were severe fetal cases. We identified pathogenic mutations in 36 of 204 (17.6%) patients. These mutations included five de novo heterozygous loss of function mutations/deletions in the PBX homeobox 1 gene (PBX1), a gene known to have a crucial role in kidney development. In contrast, the frequency of SOX17 and DSTYK variants recently reported as pathogenic in CAKUT did not indicate causality. These findings suggest that PBX1 is involved in monogenic CAKUT in humans and call into question the role of some gene variants recently reported as pathogenic in CAKUT. Targeted exome sequencing also proved to be an efficient and cost-effective strategy to identify pathogenic mutations and deletions in known CAKUT genes.


Subject(s)
DNA-Binding Proteins/genetics , Proto-Oncogene Proteins/genetics , Urogenital Abnormalities/genetics , Cohort Studies , DNA Mutational Analysis , Exome , Female , Humans , Male , Pre-B-Cell Leukemia Transcription Factor 1
6.
J Pediatr Gastroenterol Nutr ; 64(3): 378-384, 2017 03.
Article in English | MEDLINE | ID: mdl-27253662

ABSTRACT

OBJECTIVE: Early-onset inflammatory bowel diseases can result from a wide spectrum of rare mendelian disorders. Early molecular diagnosis is crucial in defining treatment and in improving life expectancy. Herein we aimed at defining the mechanism of an immunodeficiency-polyendrocrinopathy and enteropathy-X-linked (IPEX)-like disease combined with a severe immunodeficiency in 2 siblings born from distantly related parents. METHODS: Whole exome sequencing was performed on blood-extracted genomic DNA from the 2 affected children and their parents on the genomic platform of Institut IMAGINE. Candidate gene mutation was identified using the in-house software PolyWeb and confirmed by Sanger sequencing. Protein expression was determined by western blot. Flow cytometry was used to assess consequences of the mutation on lymphocyte phenotype and nuclear factor-kappa B (NF-κB) activation at diagnosis and after treatment by hematopoietic stem cell transplantation. RESULTS: We identified a homozygous missense mutation in mucosa-associated lymphoid tissue lymphoma translocation 1 gene (MALT1), which precluded protein expression. In keeping with the known function of MALT1, NF-κB-dependent lymphocyte activation was severely impaired. Moreover, there was a drastic reduction in Forkhead box P3 (FOXP3) regulatory T cells accounting for the IPEX-like phenotype. Following identification of the mutation, both children received hematopoietic stem cell transplantation, which permitted full clinical recovery. Immunological workup at 6 and 12 months after transplantation showed normal NF-κB activation and correction of regulatory T cells frequency. CONCLUSIONS: Along with FOXP3, interleukin 2 receptor alpha chain (IL2RA), and cytotoxic T-lymphocyte protein 4 precursor (CTLA-4) mutations, MALT1 deficiency should now be considered as a possible cause of IPEX-like syndrome associated with immunodeficiency that can be cured by hematopoietic stem cell transplantation.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/genetics , Genetic Diseases, X-Linked/genetics , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/deficiency , Mutation, Missense , Child , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diarrhea/diagnosis , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Markers , Homozygote , Humans , Immune System Diseases/diagnosis , Immune System Diseases/genetics , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Siblings
8.
Nat Commun ; 14(1): 3728, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349339

ABSTRACT

Loss of NBEAL2 function leads to grey platelet syndrome (GPS), a bleeding disorder characterized by macro-thrombocytopenia and α-granule-deficient platelets. A proportion of patients with GPS develop autoimmunity through an unknown mechanism, which might be related to the proteins NBEAL2 interacts with, specifically in immune cells. Here we show a comprehensive interactome of NBEAL2 in primary T cells, based on mass spectrometry identification of altogether 74 protein association partners. These include LRBA, a member of the same BEACH domain family as NBEAL2, recessive mutations of which cause autoimmunity and lymphocytic infiltration through defective CTLA-4 trafficking. Investigating the potential association between NBEAL2 and CTLA-4 signalling suggested by the mass spectrometry results, we confirm by co-immunoprecipitation that CTLA-4 and NBEAL2 interact with each other. Interestingly, NBEAL2 deficiency leads to low CTLA-4 expression in patient-derived effector T cells, while their regulatory T cells appear unaffected. Knocking-down NBEAL2 in healthy primary T cells recapitulates the low CTLA-4 expression observed in the T cells of GPS patients. Our results thus show that NBEAL2 is involved in the regulation of CTLA-4 expression in conventional T cells and provide a rationale for considering CTLA-4-immunoglobulin therapy in patients with GPS and autoimmune disease.


Subject(s)
Gray Platelet Syndrome , Humans , Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets/metabolism , Blood Proteins/genetics , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/metabolism
9.
Front Immunol ; 12: 748679, 2021.
Article in English | MEDLINE | ID: mdl-34721418

ABSTRACT

To circumvent the limitations of available preclinical models for the study of type 1 diabetes (T1D), we developed a new humanized model, the YES-RIP-hB7.1 mouse. This mouse is deficient of murine major histocompatibility complex class I and class II, the murine insulin genes, and expresses as transgenes the HLA-A*02:01 allele, the diabetes high-susceptibility HLA-DQ8A and B alleles, the human insulin gene, and the human co-stimulatory molecule B7.1 in insulin-secreting cells. It develops spontaneous T1D along with CD4+ and CD8+ T-cell responses to human preproinsulin epitopes. Most of the responses identified in these mice were validated in T1D patients. This model is amenable to characterization of hPPI-specific epitopes involved in T1D and to the identification of factors that may trigger autoimmune response to insulin-secreting cells in human T1D. It will allow evaluating peptide-based immunotherapy that may directly apply to T1D in human and complete preclinical model availability to address the issue of clinical heterogeneity of human disease.


Subject(s)
B7-1 Antigen/genetics , Diabetes Mellitus, Type 1/immunology , HLA-DQ Antigens/genetics , Insulin/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Female , H-2 Antigens/genetics , HLA-A2 Antigen/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Middle Aged , Young Adult
10.
Nat Commun ; 11(1): 5341, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087723

ABSTRACT

Autoimmunity can occur when a checkpoint of self-tolerance fails. The study of familial autoimmune diseases can reveal pathophysiological mechanisms involved in more common autoimmune diseases. Here, by whole-exome/genome sequencing we identify heterozygous, autosomal-dominant, germline loss-of-function mutations in the SOCS1 gene in ten patients from five unrelated families with early onset autoimmune manifestations. The intracellular protein SOCS1 is known to downregulate cytokine signaling by inhibiting the JAK-STAT pathway. Accordingly, patient-derived lymphocytes exhibit increased STAT activation in vitro in response to interferon-γ, IL-2 and IL-4 that is reverted by the JAK1/JAK2 inhibitor ruxolitinib. This effect is associated with a series of in vitro and in vivo immune abnormalities consistent with lymphocyte hyperactivity. Hence, SOCS1 haploinsufficiency causes a dominantly inherited predisposition to early onset autoimmune diseases related to cytokine hypersensitivity of immune cells.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/genetics , Suppressor of Cytokine Signaling 1 Protein/deficiency , Suppressor of Cytokine Signaling 1 Protein/genetics , Adolescent , Adult , Age of Onset , Autoimmune Diseases/metabolism , Child , Child, Preschool , Cytokines/metabolism , Female , Haploinsufficiency , Humans , Male , Models, Molecular , Mutation , Pedigree , STAT Transcription Factors/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/chemistry , T-Lymphocytes/immunology
11.
J Clin Invest ; 129(3): 1047-1060, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30521495

ABSTRACT

ARHGEF1 is a RhoA-specific guanine nucleotide exchange factor expressed in hematopoietic cells. We used whole-exome sequencing to identify compound heterozygous mutations in ARHGEF1, resulting in the loss of ARHGEF1 protein expression in 2 primary antibody-deficient siblings presenting with recurrent severe respiratory tract infections and bronchiectasis. Both ARHGEF1-deficient patients showed an abnormal B cell immunophenotype, with a deficiency in marginal zone and memory B cells and an increased frequency of transitional B cells. Furthermore, the patients' blood contained immature myeloid cells. Analysis of a mediastinal lymph node from one patient highlighted the small size of the germinal centers and an abnormally high plasma cell content. On the molecular level, T and B lymphocytes from both patients displayed low RhoA activity and low steady-state actin polymerization (even after stimulation of lysophospholipid receptors). As a consequence of disturbed regulation of the RhoA downstream target Rho-associated kinase I/II (ROCK), the patients' lymphocytes failed to efficiently restrain AKT phosphorylation. Enforced ARHGEF1 expression or drug-induced activation of RhoA in the patients' cells corrected the impaired actin polymerization and AKT regulation. Our results indicate that ARHGEF1 activity in human lymphocytes is involved in controlling actin cytoskeleton dynamics, restraining PI3K/AKT signaling, and confining B lymphocytes and myelocytes within their dedicated functional environment.


Subject(s)
B-Lymphocytes , Primary Immunodeficiency Diseases , Signal Transduction , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Female , Germinal Center/immunology , Germinal Center/pathology , Humans , Immunologic Memory/genetics , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/immunology , Siblings , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , rho-Associated Kinases/genetics , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/genetics
12.
EMBO Mol Med ; 10(4)2018 04.
Article in English | MEDLINE | ID: mdl-29567797

ABSTRACT

Herein, we report the first identification of biallelic-inherited mutations in ALPI as a Mendelian cause of inflammatory bowel disease in two unrelated patients. ALPI encodes for intestinal phosphatase alkaline, a brush border metalloenzyme that hydrolyses phosphate from the lipid A moiety of lipopolysaccharides and thereby drastically reduces Toll-like receptor 4 agonist activity. Prediction tools and structural modelling indicate that all mutations affect critical residues or inter-subunit interactions, and heterologous expression in HEK293T cells demonstrated that all ALPI mutations were loss of function. ALPI mutations impaired either stability or catalytic activity of ALPI and rendered it unable to detoxify lipopolysaccharide-dependent signalling. Furthermore, ALPI expression was reduced in patients' biopsies, and ALPI activity was undetectable in ALPI-deficient patient's stool. Our findings highlight the crucial role of ALPI in regulating host-microbiota interactions and restraining host inflammatory responses. These results indicate that ALPI mutations should be included in screening for monogenic causes of inflammatory bowel diseases and lay the groundwork for ALPI-based treatments in intestinal inflammatory disorders.


Subject(s)
Alkaline Phosphatase/metabolism , Inflammatory Bowel Diseases/metabolism , Alkaline Phosphatase/deficiency , Alkaline Phosphatase/genetics , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Homeostasis , Humans , Inflammatory Bowel Diseases/genetics , Intestines , Mutation/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL