Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Proc Natl Acad Sci U S A ; 120(22): e2211087120, 2023 05 30.
Article En | MEDLINE | ID: mdl-37216524

Mutations in genes encoding molecular chaperones can lead to chaperonopathies, but none have so far been identified causing congenital disorders of glycosylation. Here we identified two maternal half-brothers with a novel chaperonopathy, causing impaired protein O-glycosylation. The patients have a decreased activity of T-synthase (C1GALT1), an enzyme that exclusively synthesizes the T-antigen, a ubiquitous O-glycan core structure and precursor for all extended O-glycans. The T-synthase function is dependent on its specific molecular chaperone Cosmc, which is encoded by X-chromosomal C1GALT1C1. Both patients carry the hemizygous variant c.59C>A (p.Ala20Asp; A20D-Cosmc) in C1GALT1C1. They exhibit developmental delay, immunodeficiency, short stature, thrombocytopenia, and acute kidney injury (AKI) resembling atypical hemolytic uremic syndrome. Their heterozygous mother and maternal grandmother show an attenuated phenotype with skewed X-inactivation in blood. AKI in the male patients proved fully responsive to treatment with the complement inhibitor Eculizumab. This germline variant occurs within the transmembrane domain of Cosmc, resulting in dramatically reduced expression of the Cosmc protein. Although A20D-Cosmc is functional, its decreased expression, though in a cell or tissue-specific manner, causes a large reduction of T-synthase protein and activity, which accordingly leads to expression of varied amounts of pathological Tn-antigen (GalNAcα1-O-Ser/Thr/Tyr) on multiple glycoproteins. Transient transfection of patient lymphoblastoid cells with wild-type C1GALT1C1 partially rescued the T-synthase and glycosylation defect. Interestingly, all four affected individuals have high levels of galactose-deficient IgA1 in sera. These results demonstrate that the A20D-Cosmc mutation defines a novel O-glycan chaperonopathy and causes the altered O-glycosylation status in these patients.


Acute Kidney Injury , Molecular Chaperones , Male , Humans , Molecular Chaperones/metabolism , Mutation , Polysaccharides/metabolism , Germ Cells/metabolism
2.
J Clin Immunol ; 42(6): 1310-1320, 2022 08.
Article En | MEDLINE | ID: mdl-35670985

BACKGROUND: Deoxyribonuclease 1 like 3 (DNASE1L3) is a secreted enzyme that has been shown to digest the extracellular chromatin derived from apoptotic bodies, and DNASE1L3 pathogenic variants have been associated with a lupus phenotype. It is unclear whether interferon signaling is sustained in DNASE1L3 deficiency in humans. OBJECTIVES: To explore interferon signaling in DNASE1L3 deficient patients. To depict the characteristic features of DNASE1L3 deficiencies in human. METHODS: We identified, characterized, and analyzed five new patients carrying biallelic DNASE1L3 variations. Whole or targeted exome and/or Sanger sequencing was performed to detect pathogenic variations in five juvenile systemic erythematosus lupus (jSLE) patients. We measured interferon-stimulated gene (ISG) expression in all patients. We performed a systematic review of all published cases available from its first description in 2011 to March 24th 2022. RESULTS: We identified five new patients carrying biallelic DNASE1L3 pathogenic variations, including three previously unreported mutations. Contrary to canonical type I interferonopathies, we noticed a transient increase of ISGs in blood, which returned to normal with disease remission. Disease in one patient was characterized by lupus nephritis and skin lesions, while four others exhibited hypocomplementemic urticarial vasculitis syndrome. The fourth patient presented also with early-onset inflammatory bowel disease. Reviewing previous reports, we identified 35 additional patients with DNASE1L3 deficiency which was associated with a significant risk of lupus nephritis and a poor outcome together with the presence of anti-neutrophil cytoplasmic antibodies (ANCA). Lung lesions were reported in 6/35 patients. CONCLUSIONS: DNASE1L3 deficiencies are associated with a broad phenotype including frequently lupus nephritis and hypocomplementemic urticarial vasculitis with positive ANCA and rarely, alveolar hemorrhages and inflammatory bowel disease. This report shows that interferon production is transient contrary to anomalies of intracellular DNA sensing and signaling observed in Aicardi-Goutières syndrome or STING-associated vasculitis in infancy (SAVI).


Endodeoxyribonucleases , Inflammatory Bowel Diseases , Interferon Type I , Lupus Erythematosus, Systemic , Lupus Nephritis , Vasculitis , Antibodies, Antineutrophil Cytoplasmic/genetics , Chromatin , DNA , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Humans , Interferon Type I/genetics , Interferons , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/diagnosis , Lupus Nephritis/genetics , Phenotype , Vasculitis/diagnosis
3.
Brain ; 144(9): 2659-2669, 2021 10 22.
Article En | MEDLINE | ID: mdl-34415322

Phosphoinositides are lipids that play a critical role in processes such as cellular signalling, ion channel activity and membrane trafficking. When mutated, several genes that encode proteins that participate in the metabolism of these lipids give rise to neurological or developmental phenotypes. PI4KA is a phosphoinositide kinase that is highly expressed in the brain and is essential for life. Here we used whole exome or genome sequencing to identify 10 unrelated patients harbouring biallelic variants in PI4KA that caused a spectrum of conditions ranging from severe global neurodevelopmental delay with hypomyelination and developmental brain abnormalities to pure spastic paraplegia. Some patients presented immunological deficits or genito-urinary abnormalities. Functional analyses by western blotting and immunofluorescence showed decreased PI4KA levels in the patients' fibroblasts. Immunofluorescence and targeted lipidomics indicated that PI4KA activity was diminished in fibroblasts and peripheral blood mononuclear cells. In conclusion, we report a novel severe metabolic disorder caused by PI4KA malfunction, highlighting the importance of phosphoinositide signalling in human brain development and the myelin sheath.


Alleles , Genetic Variation/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Minor Histocompatibility Antigens/genetics , Neurodevelopmental Disorders/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Hereditary Central Nervous System Demyelinating Diseases/diagnostic imaging , Humans , Infant , Infant, Newborn , Leukocytes, Mononuclear/physiology , Male , Neurodevelopmental Disorders/diagnostic imaging , Pedigree
4.
Sci Immunol ; 6(59)2021 05 25.
Article En | MEDLINE | ID: mdl-34035116

Multiple Inflammatory Syndrome in Children (MIS-C) is a delayed and severe complication of SARS-CoV-2 infection that strikes previously healthy children. As MIS-C combines clinical features of Kawasaki disease and Toxic Shock Syndrome (TSS), we aimed to compare the immunological profile of pediatric patients with these different conditions. We analyzed blood cytokine expression, and the T cell repertoire and phenotype in 36 MIS-C cases, which were compared to 16 KD, 58 TSS, and 42 COVID-19 cases. We observed an increase of serum inflammatory cytokines (IL-6, IL-10, IL-18, TNF-α, IFNγ, CD25s, MCP1, IL-1RA) in MIS-C, TSS and KD, contrasting with low expression of HLA-DR in monocytes. We detected a specific expansion of activated T cells expressing the Vß21.3 T cell receptor ß chain variable region in both CD4 and CD8 subsets in 75% of MIS-C patients and not in any patient with TSS, KD, or acute COVID-19; this correlated with the cytokine storm detected. The T cell repertoire returned to baseline within weeks after MIS-C resolution. Vß21.3+ T cells from MIS-C patients expressed high levels of HLA-DR, CD38 and CX3CR1 but had weak responses to SARS-CoV-2 peptides in vitro. Consistently, the T cell expansion was not associated with specific classical HLA alleles. Thus, our data suggested that MIS-C is characterized by a polyclonal Vß21.3 T cell expansion not directed against SARS-CoV-2 antigenic peptides, which is not seen in KD, TSS and acute COVID-19.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19/pathology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Adult , Child , Child, Preschool , Cytokines/blood , HLA-DR Antigens/immunology , Humans , Lymphocyte Activation/immunology , SARS-CoV-2/immunology
5.
Nat Commun ; 11(1): 6182, 2020 12 03.
Article En | MEDLINE | ID: mdl-33273464

Upon sensing cytosolic DNA, the enzyme cGAS induces innate immune responses that underpin anti-microbial defenses and certain autoimmune diseases. Missense mutations of PRKDC encoding the DNA-dependent protein kinase (DNA-PK) catalytic subunit (DNA-PKcs) are associated with autoimmune diseases, yet how DNA-PK deficiency leads to increased immune responses remains poorly understood. In this study, we report that DNA-PK phosphorylates cGAS and suppresses its enzymatic activity. DNA-PK deficiency reduces cGAS phosphorylation and promotes antiviral innate immune responses, thereby potently restricting viral replication. Moreover, cells isolated from DNA-PKcs-deficient mice or patients carrying PRKDC missense mutations exhibit an inflammatory gene expression signature. This study provides a rational explanation for the autoimmunity of patients with missense mutations of PRKDC, and suggests that cGAS-mediated immune signaling is a potential target for therapeutic interventions.


Antiviral Agents/metabolism , DNA-Activated Protein Kinase/deficiency , Immunity, Innate , Nucleotidyltransferases/metabolism , Animals , Cell Line , Chromones/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Mice, Inbred C57BL , Morpholines/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , RNA, Guide, Kinetoplastida/metabolism , Signal Transduction/drug effects , Simplexvirus/drug effects , Simplexvirus/physiology , THP-1 Cells , Vesiculovirus/drug effects , Vesiculovirus/physiology , Virus Replication/drug effects
6.
Nat Commun ; 11(1): 5341, 2020 10 21.
Article En | MEDLINE | ID: mdl-33087723

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.


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
7.
Lancet Rheumatol ; 2(2): e99-e109, 2020 Feb.
Article En | MEDLINE | ID: mdl-38263665

BACKGROUND: Systemic lupus erythematosus (SLE) is a rare immunological disorder and genetic factors are considered important in its causation. Monogenic lupus has been associated with around 30 genotypes in humans and 60 in mice, while genome-wide association studies have identified more than 90 risk loci. We aimed to analyse the contribution of rare and predicted pathogenic gene variants in a population of unselected cases of childhood-onset SLE. METHODS: For this genetic panel analysis we designed a next-generation sequencing panel comprising 147 genes, including all known lupus-causing genes in humans, and potentially lupus-causing genes identified through GWAS and animal models. We screened 117 probands fulfilling American College of Rheumatology (ACR) criteria for SLE, ascertained through British and French cohorts of childhood-onset SLE, and compared these data with those of 791 ethnically matched controls from the 1000 Genomes Project and 574 controls from the FREX Consortium. FINDINGS: After filtering, mendelian genotypes were confirmed in eight probands, involving variants in C1QA, C1QC, C2, DNASE1L3, and IKZF1. Seven additional patients carried heterozygous variants in complement or type I interferon-associated autosomal recessive genes, with decreased concentrations of the encoded proteins C3 and C9 recorded in two patients. Rare variants that were predicted to be damaging were significantly enriched in the childhood-onset SLE cohort compared with controls; 25% of SLE probands versus 5% of controls were identified to harbour at least one rare, predicted damaging variant (p=2·98 × 10-11). Inborn errors of immunity were estimated to account for 7% of cases of childhood-onset SLE, with defects in innate immunity representing the main monogenic contribution. INTERPRETATION: An accumulation of rare variants that are predicted to be damaging in SLE-associated genes might contribute to disease expression and clinical heterogeneity. FUNDING: European Research Council.

8.
J Inherit Metab Dis ; 40(3): 415-422, 2017 05.
Article En | MEDLINE | ID: mdl-28255778

BACKGROUND: Mitochondrial acetoacetyl-CoA thiolase (T2) deficiency affects ketone body and isoleucine catabolism. Neurological impairment may occur secondary to ketoacidotic episodes. However, we observed neuromotor abnormalities without ketoacidotic events in two T2-deficient families. We hypothesized that the neurological signs were related to the genetic defect and may occur independently of ketoacidotic episodes. We therefore conducted a retrospective review on a French T2-deficient patient series searching for neuromotor impairment. METHODS: In total, 26 cases were retrospectively analysed for clinical, biological and neuroimaging data. RESULTS: Neurological findings were observed for 6/26 (23%) patients. Among these, two had never experienced ketoacidotic episodes, though they developed extrapyramidal signs with putamen involvement. Two of the other four patients developed neurological abnormalities before the first ketoacidotic crisis, with putamen involvement in one case. The third patient developed extrapyramidal symptoms more than 10 years after the initial decompensation with globus pallidus involvement. The last patient developed extrapyramidal signs immediately after a severe ketoacidotic crisis with putaminal lesions. CONCLUSIONS: Most T2-deficient patients achieved normal neurodevelopment. However, on account of the role of T2 in isoleucine catabolism, these patients are potentially exposed to accumulation of toxic isoleucine-derived metabolites, which may contribute to neurological impairment. Our findings confirm previous observations that neurological symptoms in T2 deficiency may occur unrelated to ketoacidosis. The role of protein restriction as a preventive measure against neurological symptoms could not be established in this study and deserves further evaluation. Long-term follow-up data on children diagnosed by newborn screening may clarify the pathogenesis of this neurometabolic association.


Acetyl-CoA C-Acetyltransferase/deficiency , Acetyl-CoA C-Acyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Basal Ganglia/metabolism , Ketosis/metabolism , Mitochondria/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Isoleucine/metabolism , Ketone Bodies/metabolism , Male , Neonatal Screening/methods , Retrospective Studies , Young Adult
9.
Joint Bone Spine ; 84(5): 589-593, 2017 Oct.
Article En | MEDLINE | ID: mdl-28039062

OBJECTIVE: Studies of early-onset systemic lupus erythematosus (SLE) have identified monogenic forms of the disease. The primary objective of this study was to compare the clinical and laboratory features of the first patients included in the GENIAL/LUMUGENE cohort to those reported in previous publications. The secondary objective was to determine whether subgroups with a distinctive pattern of clinical and biological features are seen in predominantly genetic forms of SLE. METHODS: GENIAL/LUMUGENE is a French nationwide study of the clinical, immunological, and genetic features of juvenile-onset SLE (clinicaltrials.gov #NCT01992666). Clinical and laboratory data from the first 64 patients younger than 18 years who were included in the first part of the study were collected retrospectively. Predefined criteria were used to divide the patients into three subgroups: syndromic SLE (n=10) and familial SLE (n=12) - both presumed to have a strong genetic component - and other forms of early-onset SLE (n=42). RESULTS: The predefined criteria for identifying subgroups based on knowledge of the clinical and epidemiological features of monogenic SLE showed a significantly younger age at onset in syndromic SLE (P<0.05) and a lower frequency of joint manifestations in familial SLE. CONCLUSIONS: In this study, clinical and epidemiological data alone failed to identify a specific patient subgroup characterized by the same disease presentation or progression. This result may be related to the small sample size or indicate marked heterogeneity of juvenile-onset SLE. Genetic studies using new sequencing techniques in these patients might identify genetic factors responsible for marked phenotypic variability.


Genetic Predisposition to Disease/epidemiology , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/genetics , Phenotype , Adolescent , Age Factors , Age of Onset , Child , Child, Preschool , Cohort Studies , Disease Progression , Female , Follow-Up Studies , Humans , Incidence , Lupus Erythematosus, Systemic/epidemiology , Male , Retrospective Studies , Risk Assessment , Severity of Illness Index , Sex Factors
10.
J Exp Med ; 213(6): 1011-28, 2016 05 30.
Article En | MEDLINE | ID: mdl-27185855

Inherited bone marrow failure syndromes are human conditions in which one or several cell lineages of the hemopoietic system are affected. They are present at birth or may develop progressively. They are sometimes accompanied by other developmental anomalies. Three main molecular causes have been recognized to result in bone marrow failure syndromes: (1) defects in the Fanconi anemia (FA)/BRCA DNA repair pathway, (2) defects in telomere maintenance, and (3) abnormal ribosome biogenesis. We analyzed a patient with mild bone marrow failure and microcephaly who did not present with the typical FA phenotype. Cells from this patient showed increased sensitivity to ionizing radiations and phleomycin, attesting to a probable DNA double strand break (dsb) repair defect. Linkage analysis and whole exome sequencing revealed a homozygous nonsense mutation in the ERCC6L2 gene. We identified a new ERCC6L2 alternative transcript encoding the DNA repair factor Hebo, which is critical for complementation of the patient's DNAdsb repair defect. Sequence analysis revealed three structured regions within Hebo: a TUDOR domain, an adenosine triphosphatase domain, and a new domain, HEBO, specifically present in Hebo direct orthologues. Hebo is ubiquitously expressed, localized in the nucleus, and rapidly recruited to DNAdsb's in an NBS1-dependent manner.


Bone Marrow Diseases , Cell Nucleus , Codon, Nonsense , DNA Helicases , Homozygote , Microcephaly , Adolescent , Bone Marrow Diseases/genetics , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Helicases/biosynthesis , DNA Helicases/genetics , Female , Gene Expression Regulation , Genetic Linkage , High-Throughput Nucleotide Sequencing , Humans , Male , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains
11.
PLoS Genet ; 12(3): e1005894, 2016 Mar.
Article En | MEDLINE | ID: mdl-26967905

Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, >500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of >1200 ciliary genes. NEK8/NPHP9 mutations were identified in five cases with severe overlapping phenotypes including renal cystic dysplasia/hypodysplasia, situs inversus, cardiopathy with hypertrophic septum and bile duct paucity. These cases highlight a genotype-phenotype correlation, with missense and nonsense mutations associated with hypodysplasia and enlarged cystic organs, respectively. Functional analyses of NEK8 mutations in patient fibroblasts and mIMCD3 cells showed that these mutations differentially affect ciliogenesis, proliferation/apoptosis/DNA damage response, as well as epithelial morphogenesis. Notably, missense mutations exacerbated some of the defects due to NEK8 loss of function, highlighting their likely gain-of-function effect. We also showed that NEK8 missense and loss-of-function mutations differentially affect the regulation of the main Hippo signaling effector, YAP, as well as the expression of its target genes in patient fibroblasts and renal cells. YAP imbalance was also observed in enlarged spheroids of Nek8-invalidated renal epithelial cells grown in 3D culture, as well as in cystic kidneys of Jck mice. Moreover, co-injection of nek8 MO with WT or mutated NEK8-GFP RNA in zebrafish embryos led to shortened dorsally curved body axis, similar to embryos injected with human YAP RNA. Finally, treatment with Verteporfin, an inhibitor of YAP transcriptional activity, partially rescued the 3D spheroid defects of Nek8-invalidated cells and the abnormalities of NEK8-overexpressing zebrafish embryos. Altogether, our study demonstrates that NEK8 human mutations cause major organ developmental defects due to altered ciliogenesis and cell differentiation/proliferation through deregulation of the Hippo pathway.


Adaptor Proteins, Signal Transducing/genetics , Cilia/genetics , Phosphoproteins/genetics , Polycystic Kidney Diseases/genetics , Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Cell Differentiation/genetics , Cilia/pathology , Female , Genetic Association Studies , Humans , Kidney/metabolism , Kidney/pathology , Mice , Morphogenesis/genetics , Mutation , NIMA-Related Kinases , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/biosynthesis , Polycystic Kidney Diseases/pathology , Porphyrins/administration & dosage , Signal Transduction , Transcription Factors , Verteporfin , YAP-Signaling Proteins , Zebrafish
12.
Int J Radiat Oncol Biol Phys ; 94(3): 450-60, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26867874

PURPOSE: Whereas post-radiation therapy overreactions (OR) represent a clinical and societal issue, there is still no consensual radiobiological endpoint to predict clinical radiosensitivity. Since 2003, skin biopsy specimens have been collected from patients treated by radiation therapy against different tumor localizations and showing a wide range of OR. Here, we aimed to establish quantitative links between radiobiological factors and OR severity grades that would be relevant to radioresistant and genetic hyperradiosensitive cases. METHODS AND MATERIALS: Immunofluorescence experiments were performed on a collection of skin fibroblasts from 12 radioresistant, 5 hyperradiosensitive, and 100 OR patients irradiated at 2 Gy. The numbers of micronuclei, γH2AX, and pATM foci that reflect different steps of DNA double-strand breaks (DSB) recognition and repair were assessed from 10 minutes to 24 hours after irradiation and plotted against the severity grades established by the Common Terminology Criteria for Adverse Events and the Radiation Therapy Oncology Group. RESULTS: OR patients did not necessarily show a gross DSB repair defect but a systematic delay in the nucleoshuttling of the ATM protein required for complete DSB recognition. Among the radiobiological factors, the maximal number of pATM foci provided the best discrimination among OR patients and a significant correlation with each OR severity grade, independently of tumor localization and of the early or late nature of reactions. CONCLUSIONS: Our results are consistent with a general classification of human radiosensitivity based on 3 groups: radioresistance (group I); moderate radiosensitivity caused by delay of nucleoshuttling of ATM, which includes OR patients (group II); and hyperradiosensitivity caused by a gross DSB repair defect, which includes fatal cases (group III).


Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Nucleus/metabolism , DNA Breaks, Double-Stranded , Histones/metabolism , Radiation Injuries/classification , Radiation Tolerance/physiology , Skin/radiation effects , Analysis of Variance , Ataxia Telangiectasia Mutated Proteins/genetics , Biopsy , Cell Line , DNA Repair , Fibroblasts/radiation effects , Humans , Micronucleus Tests/methods , Phosphorylation , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radiation Tolerance/genetics , Skin/pathology , Time Factors
13.
DNA Cell Biol ; 34(7): 489-96, 2015 Jul.
Article En | MEDLINE | ID: mdl-25849606

Molecular diagnosis of cardiomyopathies remains difficult not only because of the large number of causative genes and the high rate of private mutations but also due to the large number of unclassified variants (UVs) found in patients' DNA. This study reports the functional splicing impact of nine novel genomic variations previously identified in unrelated patients with cardiomyopathies. To identify splice variants among these UVs, a combination of in silico and in vitro hybrid minigene tools was used as transcript is not available. Using this two-step approach, these UVs were reclassified as splicing mutations (MYBPC3-c.655-25A>G, MYBPC3-c.1790G>A (p.Arg597Gln), MYBPC3-c.2414-36G>T) or as mutations with a majority of abnormally spliced transcripts (MYBPC3-c.1182C>A, TNNT2-c.460G>A (p.Glu154Lys), and TNNT2-c.822-3C>A) or as variations with a weak splicing effect (TNNT2-c.1000-38C>A). For the two remaining variations in intron 11 of the TNNT2 gene in the vicinity of the acceptor splice site (c.571-7G>A, c.571-29G>A), a minigene assay was inconclusive as exon 12 is neither recognized as an exon by HeLa nor by H9c2 cells. Our study highlights the importance of the combined use of in silico and in vitro splicing assays to improve the prediction of the functional splicing impact of identified genetic variants if the RNA sample from the patient is not easily available.


Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Alternative Splicing , Animals , Carrier Proteins/genetics , HeLa Cells , Humans , Point Mutation , Polymorphism, Single Nucleotide , RNA Splice Sites , Rats , Sequence Analysis, DNA , Troponin T/genetics
14.
J Allergy Clin Immunol ; 135(6): 1578-88.e5, 2015 Jun.
Article En | MEDLINE | ID: mdl-25842288

BACKGROUND: PRKDC encodes for DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a kinase that forms part of a complex (DNA-dependent protein kinase [DNA-PK]) crucial for DNA double-strand break repair and V(D)J recombination. In mice DNA-PK also interacts with the transcription factor autoimmune regulator (AIRE) to promote central T-cell tolerance. OBJECTIVE: We sought to understand the causes of an inflammatory disease with granuloma and autoimmunity associated with decreasing T- and B-cell counts over time that had been diagnosed in 2 unrelated patients. METHODS: Genetic, molecular, and functional analyses were performed to characterize an inflammatory disease evocative of a combined immunodeficiency. RESULTS: We identified PRKDC mutations in both patients. These patients exhibited a defect in DNA double-strand break repair and V(D)J recombination. Whole-blood mRNA analysis revealed a strong interferon signature. On activation, memory T cells displayed a skewed cytokine response typical of TH2 and TH1 but not TH17. Moreover, mutated DNA-PKcs did not promote AIRE-dependent transcription of peripheral tissue antigens in vitro. The latter defect correlated in vivo with production of anti-calcium-sensing receptor autoantibodies, which are typically found in AIRE-deficient patients. In addition, 9 months after bone marrow transplantation, patient 1 had Hashimoto thyroiditis, suggesting that organ-specific autoimmunity might be linked to nonhematopoietic cells, such as AIRE-expressing thymic epithelial cells. CONCLUSION: Deficiency of DNA-PKcs, a key AIRE partner, can present as an inflammatory disease with organ-specific autoimmunity, suggesting a role for DNA-PKcs in regulating autoimmune responses and maintaining AIRE-dependent tolerance in human subjects.


DNA-Activated Protein Kinase/genetics , Granuloma/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , Nuclear Proteins/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Adolescent , Animals , Autoantibodies/biosynthesis , Autoimmunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , DNA End-Joining Repair/immunology , DNA-Activated Protein Kinase/deficiency , DNA-Activated Protein Kinase/immunology , Female , Gene Expression Regulation , Granuloma/immunology , Granuloma/metabolism , Granuloma/pathology , Humans , Immune Tolerance , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Male , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Transcription Factors/immunology , V(D)J Recombination/immunology , Young Adult , AIRE Protein
15.
Neurology ; 81(17): 1523-30, 2013 Oct 22.
Article En | MEDLINE | ID: mdl-24027061

OBJECTIVE: To investigate whether mutations in the SURF1 gene are a cause of Charcot-Marie-Tooth (CMT) disease. METHODS: We describe 2 patients from a consanguineous family with demyelinating autosomal recessive CMT disease (CMT4) associated with the homozygous splice site mutation c.107-2A>G in the SURF1 gene, encoding an assembly factor of the mitochondrial respiratory chain complex IV. This observation led us to hypothesize that mutations in SURF1 might be an unrecognized cause of CMT4, and we investigated SURF1 in a total of 40 unrelated patients with CMT4 after exclusion of mutations in known CMT4 genes. The functional impact of c.107-2A>G on splicing, amount of SURF1 protein, and on complex IV activity and assembly was analyzed. RESULTS: Another patient with CMT4 was found to harbor 2 additional SURF1 mutations. All 3 patients with SURF1-associated CMT4 presented with severe childhood-onset neuropathy, motor nerve conduction velocities <25 m/s, and lactic acidosis. Two patients had brain MRI abnormalities, including putaminal and periaqueductal lesions, and developed cerebellar ataxia years after polyneuropathy. The c.107-2A>G mutation produced no normally spliced transcript, leading to SURF1 absence. However, complex IV remained partially functional in muscle and fibroblasts. CONCLUSIONS: We found SURF1 mutations in 5% of families (2/41) presenting with CMT4. SURF1 should be systematically screened in patients with childhood-onset severe demyelinating neuropathy and additional features such as lactic acidosis, brain MRI abnormalities, and cerebellar ataxia developing years after polyneuropathy.


Charcot-Marie-Tooth Disease/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Adult , Age of Onset , Charcot-Marie-Tooth Disease/pathology , Child, Preschool , Consanguinity , Female , Homozygote , Humans , Male , Membrane Proteins/deficiency , Middle Aged , Mitochondrial Proteins/deficiency , Mutation/genetics , Pedigree , Phenotype , RNA Splicing/genetics
16.
Arthritis Rheum ; 65(8): 2161-71, 2013 Aug.
Article En | MEDLINE | ID: mdl-23666743

OBJECTIVE: Systemic lupus erythematosus (SLE) is a prototype autoimmune disease that is assumed to occur via a complex interplay of environmental and genetic factors. Rare causes of monogenic SLE have been described, providing unique insights into fundamental mechanisms of immune tolerance. The aim of this study was to identify the cause of an autosomal-recessive form of SLE. METHODS: We studied 3 siblings with juvenile-onset SLE from 1 consanguineous kindred and used next-generation sequencing to identify mutations in the disease-associated gene. We performed extensive biochemical, immunologic, and functional assays to assess the impact of the identified mutations on B cell biology. RESULTS: We identified a homozygous missense mutation in PRKCD, encoding protein kinase δ (PKCδ), in all 3 affected siblings. Mutation of PRKCD resulted in reduced expression and activity of the encoded protein PKCδ (involved in the deletion of autoreactive B cells), leading to resistance to B cell receptor- and calcium-dependent apoptosis and increased B cell proliferation. Thus, as for mice deficient in PKCδ, which exhibit an SLE phenotype and B cell expansion, we observed an increased number of immature B cells in the affected family members and a developmental shift toward naive B cells with an immature phenotype. CONCLUSION: Our findings indicate that PKCδ is crucial in regulating B cell tolerance and preventing self-reactivity in humans, and that PKCδ deficiency represents a novel genetic defect of apoptosis leading to SLE.


Apoptosis , B-Lymphocytes/pathology , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/genetics , Mutation, Missense , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/genetics , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Child , Female , Genetic Variation , Homozygote , Humans , Hyperplasia , Immune Tolerance , Lupus Erythematosus, Systemic/pathology , Male , Polymorphism, Single Nucleotide , Protein Kinase C-delta/immunology , Young Adult
17.
Antimicrob Agents Chemother ; 56(7): 3732-8, 2012 Jul.
Article En | MEDLINE | ID: mdl-22526305

In bone marrow transplantation, the efficacy of ganciclovir in cytomegalovirus (CMV) disease treatment or prophylaxis remains partial. Because its hematological toxicity is dose limiting, optimization of the dosing schedule is required to increase its therapeutic index. The goal of our study was to describe the influence of the ganciclovir concentration and duration of exposure on cell survival and antiviral efficacy. The study was carried out in vitro on cultures of lymphoblastoid cells infected or not with the CMV AD169 reference strain and exposed to ganciclovir at different concentrations for 1, 2, 7, or 14 days. The data were analyzed by a mathematical model that allowed a quantitative characterization of ganciclovir pharmacodynamics and its variability. Simulations of the model were undertaken to determine the optimal concentration profile for maximizing the ganciclovir therapeutic index. Ganciclovir had very little toxic and antiviral effect, even at 20 mg liter(-1), when the duration of exposure was ≤ 7 days. A biologically significant effect was observed only with a 14-day exposure. Complete inhibition of viral replication was obtained at 20 mg liter(-1). The utility function, assuming equal weights for antiviral effect and toxicity, showed that maximal utility was reached around 10 mg liter(-1). The optimal ganciclovir concentration profile consisted of maintaining the concentration at 20 mg liter(-1) at the intervals 0 to 2 days and 7.58 to 9.58 days and a null concentration at other times. This optimal profile could be obtained by intravenous (i.v.) ganciclovir at 10 mg/kg of body weight twice daily (b.i.d.) at days 1, 2, 8.5, and 9.5 in stem cell transplant patients with normal renal function.


Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Cytomegalovirus Infections/virology , Cytomegalovirus/drug effects , Ganciclovir/adverse effects , Ganciclovir/pharmacology , Antiviral Agents/pharmacokinetics , Cell Survival/drug effects , Cells, Cultured , Ganciclovir/pharmacokinetics , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Models, Theoretical
18.
J Med Genet ; 49(2): 146-50, 2012 Feb.
Article En | MEDLINE | ID: mdl-22187496

BACKGROUND: The ANT1 gene, encoding ADP/ATP translocase 1, was investigated in an adult patient with an autosomal recessive mitochondrial disorder characterised by congenital cataracts, hypertrophic cardiomyopathy, myopathy and lactic acidosis. METHODS AND RESULTS: ANT1 sequencing showed that the patient was homozygous for a new nucleotide variation, c.111+1G→A, abolishing the invariant GT splice donor site of intron 1. The ANT1 transcript was undetectable in both muscle and skin fibroblasts. A markedly abnormal metabolic profile was found, and skeletal muscle showed a dramatic proliferation of abnormal mitochondria, increased mitochondrial mass, and multiple mitochondrial DNA deletions. No compensating increase in the transcript level of the ANT3 gene, which encodes the human ubiquitous isoform of the ADP/ATP translocase, was observed. The patient's heterozygous mother had normal clinical, biochemical and pathological features. CONCLUSIONS: Complete loss of expression of the ANT1 gene causes a clinical syndrome mainly characterised by cardiomyopathy and myopathy. This report expands the clinical spectrum of ANT1-related human diseases, and emphasises the crucial role of the mitochondrial ADP/ATP carriers in muscle function and pathophysiology of human myopathies.


Adenine Nucleotide Translocator 1/genetics , Cardiomyopathy, Hypertrophic/genetics , Mitochondrial Myopathies/genetics , Adenine Nucleotide Translocator 3/genetics , Adult , Base Sequence , Cardiomyopathy, Hypertrophic/diagnosis , Cells, Cultured , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Exons , Female , Gene Expression , Humans , Magnetic Resonance Imaging , Mitochondrial Myopathies/diagnosis , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Mutation , Neuroimaging , Pedigree , Young Adult
19.
Hum Mol Genet ; 20(18): 3653-66, 2011 Sep 15.
Article En | MEDLINE | ID: mdl-21685203

By providing access to affected neurons, human induced pluripotent stem cells (iPSc) offer a unique opportunity to model human neurodegenerative diseases. We generated human iPSc from the skin fibroblasts of children with mucopolysaccharidosis type IIIB. In this fatal lysosomal storage disease, defective α-N-acetylglucosaminidase interrupts the degradation of heparan sulfate (HS) proteoglycans and induces cell disorders predominating in the central nervous system, causing relentless progression toward severe mental retardation. Partially digested proteoglycans, which affect fibroblast growth factor signaling, accumulated in patient cells. They impaired isolation of emerging iPSc unless exogenous supply of the missing enzyme cleared storage and restored cell proliferation. After several passages, patient iPSc starved of an exogenous enzyme continued to proliferate in the presence of fibroblast growth factor despite HS accumulation. Survival and neural differentiation of patient iPSc were comparable with unaffected controls. Whereas cell pathology was modest in floating neurosphere cultures, undifferentiated patient iPSc and their neuronal progeny expressed cell disorders consisting of storage vesicles and severe disorganization of Golgi ribbons associated with modified expression of the Golgi matrix protein GM130. Gene expression profiling in neural stem cells pointed to alterations of extracellular matrix constituents and cell-matrix interactions, whereas genes associated with lysosome or Golgi apparatus functions were downregulated. Taken together, these results suggest defective responses of patient undifferentiated stem cells and neurons to environmental cues, which possibly affect Golgi organization, cell migration and neuritogenesis. This could have potential consequences on post-natal neurological development, once HS proteoglycan accumulation becomes prominent in the affected child brain.


Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Lysosomes/metabolism , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/physiopathology , Neurons/cytology , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/metabolism , Lysosomes/enzymology , Male , Models, Biological , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mutation , Neurons/enzymology , Neurons/metabolism
20.
Mitochondrion ; 11(1): 223-7, 2011 Jan.
Article En | MEDLINE | ID: mdl-20691285

The POLG genes were sequenced in two unrelated patients presenting with Alpers syndrome. The novel c.3626_3629dupGATA and the c.3643+2T>C alleles were associated in trans with p.A467T and p.[W748S;E1143G], respectively. POLG transcripts from skin fibroblasts showed complete exon 22 skipping for patient 2, but surprisingly partial exon 22 skipping from the c.3626_3629dupGATA for patient 1. The creation of a putative exonic splicing silencer could be responsible for the splicing anomaly observed in patient 1. Both c.3643+2T>C and c.3626_3629dupGATA create a premature termination codon and a low polymerase γ activity in skin fibroblasts is responsible for the severe phenotype in these patients.


DNA-Directed DNA Polymerase/genetics , Diffuse Cerebral Sclerosis of Schilder/genetics , Exons/genetics , Genetic Variation , RNA Splicing , Child, Preschool , Codon, Nonsense/genetics , DNA Polymerase gamma , DNA-Directed DNA Polymerase/metabolism , Diffuse Cerebral Sclerosis of Schilder/diagnosis , Fatal Outcome , Female , Fibroblasts/metabolism , Humans , Male , Mitochondria/enzymology , Mitochondria/genetics , Mutation , Sequence Analysis, DNA
...