Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Eur J Med Genet ; 68: 104919, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38355093

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT), also known as Rendu-Osler-Weber disease, is a dominant inherited vascular disorder. The clinical diagnosis is based on the Curaçao criteria and pathogenic variants in the ENG and ACVRL1 genes are responsible for most cases of HHT. Four families with a negative targeted gene panel and selected by a multidisciplinary team were selected and whole-genome sequencing was performed according to the recommendations of the French National Plan for Genomic Medicine. Structural variations were confirmed by standard molecular cytogenetic analysis (FISH). In two families with a definite diagnosis of HHT, we identified two different paracentric inversions of chromosome 9, both disrupting the ENG gene. These inversions are considered as pathogenic and causative for the HHT phenotype of the patients. This is the first time structural variations are reported to cause HHT. As such balanced events are often missed by exon-based sequencing (panel, exome), structural variations may be an under-recognized cause of HHT. Genome sequencing for the detection of these events could be suggested for patients with a definite diagnosis of HHT and in whom no causative pathogenic variant was identified.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic , Humans , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Mutation , Endoglin/genetics , Base Sequence , Chromosomes, Human, Pair 9/genetics , Activin Receptors, Type II/genetics
2.
Angiogenesis ; 27(2): 211-227, 2024 May.
Article in English | MEDLINE | ID: mdl-38294582

ABSTRACT

Heterozygous activin receptor-like kinase 1 (ALK1) mutations are associated with two vascular diseases: hereditary hemorrhagic telangiectasia (HHT) and more rarely pulmonary arterial hypertension (PAH). Here, we aimed to understand the impact of ALK1 mutations on BMP9 and BMP10 transcriptomic responses in endothelial cells. Endothelial colony-forming cells (ECFCs) and microvascular endothelial cells (HMVECs) carrying loss of function ALK1 mutations were isolated from newborn HHT and adult PAH donors, respectively. RNA-sequencing was performed on each type of cells compared to controls following an 18 h stimulation with BMP9 or BMP10. In control ECFCs, BMP9 and BMP10 stimulations induced similar transcriptomic responses with around 800 differentially expressed genes (DEGs). ALK1-mutated ECFCs unexpectedly revealed highly similar transcriptomic profiles to controls, both at the baseline and upon stimulation, and normal activation of Smad1/5 that could not be explained by a compensation in cell-surface ALK1 level. Conversely, PAH HMVECs revealed strong transcriptional dysregulations compared to controls with > 1200 DEGs at the baseline. Consequently, because our study involved two variables, ALK1 genotype and BMP stimulation, we performed two-factor differential expression analysis and identified 44 BMP9-dysregulated genes in mutated HMVECs, but none in ECFCs. Yet, the impaired regulation of at least one hit, namely lunatic fringe (LFNG), was validated by RT-qPCR in three different ALK1-mutated endothelial models. In conclusion, ALK1 heterozygosity only modified the BMP9/BMP10 regulation of few genes, including LFNG involved in NOTCH signaling. Future studies will uncover whether dysregulations in such hits are enough to promote HHT/PAH pathogenesis, making them potential therapeutic targets, or if second hits are necessary.


Subject(s)
Pulmonary Arterial Hypertension , Telangiectasia, Hereditary Hemorrhagic , Adult , Infant, Newborn , Humans , Endothelial Cells/metabolism , Growth Differentiation Factor 2/genetics , Growth Differentiation Factor 2/metabolism , Pulmonary Arterial Hypertension/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Bone Morphogenetic Proteins/genetics , Mutation/genetics , Gene Expression Profiling , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism
3.
Rev Mal Respir ; 40(5): 391-405, 2023 May.
Article in French | MEDLINE | ID: mdl-37062633

ABSTRACT

Hereditary hemorrhagic telangiectasia, also known as Rendu-Osler - Weber disease, is a rare, autosomal dominant vascular disease, with prevalence of 1/5,000. The condition is characterized by muco-cutaneous telangiectasias, which are responsible for a hemorrhagic syndrome of variable severity, as well as arteriovenous malformations (AVMs) appearing in the lungs, the liver, and the nervous system. They can be the source of shunts, which may be associated with high morbidity (neurological ischemic stroke, brain abscess, high-output heart failure, biliary ischemia…). It is therefore crucial to establish a clinical diagnosis using the Curaçao criteria or molecular diagnosis based on genetic analysis of the ENG, ACVRL1, SMAD4 and GDF2 genes. In most cases, multidisciplinary management allows patients to have normal life expectancy. Advances in interventional radiology and better understanding of the pathophysiology of angiogenesis have resulted in improved therapeutic management. Anti-angiogenic treatments, such as bevacizumab (BVZ, an anti-VEGF antibody), have proven to be effective in cases involving bleeding complications and severe liver damage with cardiac repercussions. Other anti-angiogenic agents are currently being investigated, including tyrosine kinase inhibitors.


Subject(s)
Arteriovenous Malformations , Telangiectasia, Hereditary Hemorrhagic , Humans , Telangiectasia, Hereditary Hemorrhagic/complications , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/epidemiology , Arteriovenous Malformations/complications , Lung , Bevacizumab , Prevalence , Activin Receptors, Type II
5.
Eur J Med Genet ; 65(1): 104370, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34737116

ABSTRACT

Hereditary haemorrhagic telangiectasia (HHT) is a complex, multisystemic vascular dysplasia affecting approximately 85,000 European Citizens. In 2016, eight founding centres operating within 6 countries, set up a working group dedicated to HHT within what became the European Reference Network on Rare Multisystemic Vascular Diseases. By launch, combined experience exceeded 10,000 HHT patients, and Chairs representing 7 separate specialties provided a median of 24 years' experience in HHT. Integrated were expert patients who focused discussions on the patient experience. Following a 2016-2017 survey to capture priorities, and underpinned by more than 40 monthly meetings, and new data acquisitions, VASCERN HHT generated position statements that distinguish expert HHT care from non-expert HHT practice. Leadership was by specialists in the relevant sub-discipline(s), and 100% consensus was required amongst all clinicians before statements were published or disseminated. One major set of outputs targeted all healthcare professionals and their HHT patients, and include the new Orphanet definition; Do's and Don'ts for common situations; Outcome Measures suitable for all consultations; COVID-19; and anticoagulation. The second output set span aspects of vascular pathophysiology where greater understanding will assist organ-specific specialist clinicians to provide more informed care to HHT patients. These cover cerebral vascular malformations and screening; mucocutaneous telangiectasia and differential diagnosis; anti-angiogenic therapies; circulatory interplays between anaemia and arteriovenous malformations; and microbiological strategies to counteract loss of normal pulmonary capillary function. Overall, the integrated outputs, and documented current practices, provide frameworks for approaches that augment the health and safety of HHT patients in diverse health-care settings.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic/therapy , Disease Management , Europe , Humans , Practice Guidelines as Topic , Rare Diseases , Telangiectasia, Hereditary Hemorrhagic/diagnosis
6.
Ann. intern. med ; 173(12): 989-1001, Dec. 15, 2020.
Article in English | BIGG - GRADE guidelines | ID: biblio-1146660

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disease with an estimated prevalence of 1 in 5000 that is characterized by the presence of vascular malformations (VMs). These result in chronic bleeding, acute hemorrhage, and complications from shunting through VMs. The goal of the Second International HHT Guidelines process was to develop evidence-based consensus guidelines for the management and prevention of HHT-related symptoms and complications. The guidelines were developed using the AGREE II (Appraisal of Guidelines for Research and Evaluation II) framework and GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology. The guidelines expert panel included expert physicians (clinical and genetic) in HHT from 15 countries, guidelines methodologists, health care workers, health care administrators, patient advocacy representatives, and persons with HHT. During the preconference process, the expert panel generated clinically relevant questions in 6 priority topic areas. A systematic literature search was done in June 2019, and articles meeting a priori criteria were included to generate evidence tables, which were used as the basis for recommendation development. The expert panel subsequently convened during a guidelines conference to conduct a structured consensus process, during which recommendations reaching at least 80% consensus were discussed and approved. The expert panel generated and approved 6 new recommendations for each of the following 6 priority topic areas: epistaxis, gastrointestinal bleeding, anemia and iron deficiency, liver VMs, pediatric care, and pregnancy and delivery (36 total). The recommendations highlight new evidence in existing topics from the first International HHT Guidelines and provide guidance in 3 new areas: anemia, pediatrics, and pregnancy and delivery. These recommendations should facilitate implementation of key components of HHT care into clinical practice.


Subject(s)
Humans , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/prevention & control , Vascular Malformations/genetics , Epistaxis/prevention & control , Gastrointestinal Hemorrhage/prevention & control , Nasal Mucosa
7.
Diagn Interv Imaging ; 101(5): 289-297, 2020 May.
Article in English | MEDLINE | ID: mdl-31932228

ABSTRACT

PURPOSE: To determine the lowest suitable dose level for the detection of pulmonary arteriovenous malformation (PAVM) using a task-based image quality assessment. MATERIAL AND METHODS: A phantom was scanned using the standard chest protocol (STD) and 4 other ultra-low dose protocols (ULD) using various kVp. Raw data were reconstructed using level 5 of the hybrid iterative reconstruction algorithm (iDose4) for the STD protocol, and level 6 of iDose4 and levels 1 to 3 of model-based iterative reconstruction (IMR) for the ULD protocols. Both quantitative criteria and qualitative analysis were used to compare protocols. Noise-power-spectrum and Task-based transfer function were computed using imQuest software. The detectability-index (d') was computed for the detection of PAVM. A subjective analysis was performed by 2 chest radiologists to validate the image-quality obtained on the anthropomorphic phantom for all protocols. RESULTS: Similar d' values were found for ULD-140 using iDose4 6 compared to STD protocol. Greater d' values were found for all ULD protocols using IMR compared to STD. Subjective image quality was rated as acceptable to excellent for ULD-140 and ULD-120 for all reconstruction types, for ULD-100 and ULD-80 using IMR2, and for ULD-100 using IMR1. Image smoothing was poor for IMR3 for ULD-100 and ULD-80. Finally, the ULD-80 protocol reconstructed with IMR2 was chosen for the detection of PAVM. With this protocol, the dose (CTDIvol of 0.3mGy) was reduced by 91% compared with the STD protocol. CONCLUSION: A dose level as low as 0.3mGy reconstructed with IMR2 provides an image quality suitable for the detection of PAVM.


Subject(s)
Arteriovenous Malformations , Radiographic Image Interpretation, Computer-Assisted , Algorithms , Arteriovenous Malformations/diagnostic imaging , Humans , Phantoms, Imaging , Radiation Dosage , Tomography, X-Ray Computed
8.
Orphanet J Rare Dis ; 14(1): 210, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31462308

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is a rare vascular dysplasia resulting in visceral arteriovenous malformations and smaller mucocutaneous telangiectasia. Most patients experience recurrent nosebleeds and become anemic without iron supplementation. However, thousands may require anticoagulation for conditions such as venous thromboembolism and/or atrial fibrillation. Over decades, tolerance data has been published for almost 200 HHT-affected users of warfarin and heparins, but there are no published data for the newer direct oral anticoagulants (DOACs) in HHT. METHODS: To provide such data, a retrospective audit was conducted across the eight HHT centres of the European Reference Network for Rare Multisystemic Vascular Diseases (VASCERN), in Denmark, France, Germany, Italy, the Netherlands and the UK. RESULTS: Although HHT Centres had not specifically recommended the use of DOACs, 32 treatment episodes had been initiated by other clinicians in 28 patients reviewed at the Centres, at median age 65 years (range 30-84). Indications were for atrial fibrillation (16 treatment episodes) and venous thromboembolism (16 episodes). The 32 treatment episodes used Apixaban (n = 15), Rivaroxaban (n = 14), and Dabigatran (n = 3). HHT nosebleeds increased in severity in 24/32 treatment episodes (75%), leading to treatment discontinuation in 11 (34.4%). Treatment discontinuation was required for 4/15 (26.7%) Apixaban episodes and 7/14 (50%) Rivaroxaban episodes. By a 4 point scale of increasing severity, there was a trend for Rivaroxaban to be associated with a greater bleeding risk both including and excluding patients who had used more than one agent (age-adjusted coefficients 0.61 (95% confidence intervals 0.11, 1.20) and 0.74 (95% confidence intervals 0.12, 1.36) respectively. Associations were maintained after adjustment for gender and treatment indication. Extreme hemorrhagic responses, worse than anything experienced previously, with individual nosebleeds lasting hours requiring hospital admissions, blood transfusions and in all cases treatment discontinuation, occurred in 5/14 (35.7%) Rivaroxaban episodes compared to 3/15 (20%) Apixaban episodes and published rates of ~ 5% for warfarin and heparin. CONCLUSIONS: Currently, conventional heparin and warfarin remain first choice anticoagulants in HHT. If newer anticoagulants are considered, although study numbers are small, at this stage Apixaban appears to be associated with lesser bleeding risk than Rivaroxaban.


Subject(s)
Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Administration, Oral , Aged , Aged, 80 and over , Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Dabigatran/administration & dosage , Dabigatran/adverse effects , Dabigatran/therapeutic use , Epistaxis/drug therapy , Female , Humans , Male , Middle Aged , Pulmonary Embolism/drug therapy , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/therapeutic use , Retrospective Studies , Rivaroxaban/administration & dosage , Rivaroxaban/adverse effects , Rivaroxaban/therapeutic use , Venous Thromboembolism , Warfarin/administration & dosage , Warfarin/adverse effects , Warfarin/therapeutic use
10.
J Thromb Haemost ; 12(9): 1494-502, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25040799

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder associated with abnormal angiogenesis and disabling epistaxis. Tranexamic acid (TA) has been widely used in the treatment of these severe bleeds but with no properly designed trial. OBJECTIVES: To demonstrate the efficacy of TA in epistaxis in HHT patients and to explore its safety of use. PATIENTS/METHODS: A randomized, placebo-controlled, double-blind, cross-over trial was conducted. Participants were randomized to receive TA (3 g a day) then placebo or the opposite sequence. The main analysis compared intra-individual mean duration of epistaxis under TA vs. placebo on a log scale. The primary outcome was the mean duration of epistaxis per month, assessed with specific grids to be completed by participants. The number of epistaxis episodes was recorded as a secondary outcome. RESULTS: A total of 118 randomized patients contributed to the statistical analysis. The mean duration of epistaxis per month was significantly shorter with TA than placebo (0.19 on the log scale; SD = 0.07; P = 0.005), corresponding to a decrease of 17.3% (15.7 min) in the duration of epistaxis per month (CI 95%, 5.5-27.6). The median number of epistaxis episodes per month was 22.1 episodes in the placebo arm vs. 23.3 episodes in the TA arm. No thrombophlebitis was observed. CONCLUSIONS: In the ATERO study, we demonstrated a significant decrease in the duration of epistaxis in HHT patients taking TA. No safety issues were recorded in our cohort of patients.


Subject(s)
Antifibrinolytic Agents/therapeutic use , Epistaxis/drug therapy , Hemorrhage/drug therapy , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Tranexamic Acid/therapeutic use , Adult , Aged , Cross-Over Studies , Double-Blind Method , Europe , Female , Humans , Male , Middle Aged , Neovascularization, Pathologic , Quality of Life , Rare Diseases , Treatment Outcome , Young Adult
11.
J Intern Med ; 274(4): 351-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23772771

ABSTRACT

OBJECTIVE: Hereditary haemorrhagic telangiectasia (HHT) is a genetic disorder related to mutations in one of the coreceptors to the transforming growth factor-ß superfamily (ALK1 or endoglin). Besides the obvious vascular symptoms (epistaxis and arteriovenous malformations), patients have an unexplained high risk of severe bacterial infections. The aim of the study was to assess the main immunological functions of patients with HHT using the standard biological tests for primary immunodeficiencies. DESIGN, SETTING AND SUBJECTS: A prospective single-centre study of 42 consecutive adult patients with an established diagnosis of HHT was conducted at the National French HHT Reference Center (Lyon). Lymphocyte subpopulations and proliferation capacity, immunoglobulin levels and neutrophil and monocyte phagocytosis, oxidative burst and chemotaxis were assessed. RESULTS: Innate immunity was not altered in patients with HHT. With regard to adaptive immunity, significant changes were seen in immunological parameters: primarily, a lymphopenia in patients with HHT compared with healthy control subjects affecting mean CD4 (642 cells µL(-1) vs. 832 cells µL(-1) , P < 0.001), CD8 (295 cells µL(-1) vs. 501 cells µL(-1) , P < 0.0001) and natural killer (NK) cells (169 cells µL(-1) vs. 221 cells µL(-1) , P < 0.01), associated with increased levels of immunoglobulins G and A. This lymphopenia mainly concerned naïve T cells. Proliferation capacities of lymphocytes were normal. Lymphopenic patients had a higher frequency of iron supplementation but no increase in infection rate. Lower levels of immunoglobulin M and a higher rate of pulmonary arteriovenous malformations were found amongst patients with a history of severe infection. CONCLUSIONS: Patients with HHT exhibit immunological abnormalities including T CD4, T CD8 and NK cell lymphopenia and increased levels of immunoglobulins G and A. The observed low level of immunoglobulin M requires further investigation to determine whether it is a specific risk factor for infection in HHT.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hypergammaglobulinemia/etiology , Lymphopenia/etiology , Telangiectasia, Hereditary Hemorrhagic/complications , Adult , Aged , Aged, 80 and over , Female , Humans , Hypergammaglobulinemia/immunology , Immunity, Innate/genetics , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Lymphopenia/immunology , Male , Middle Aged , Prospective Studies , Risk Factors , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/immunology , Young Adult
12.
J Gynecol Obstet Biol Reprod (Paris) ; 42(2): 159-65, 2013 Apr.
Article in French | MEDLINE | ID: mdl-23040267

ABSTRACT

INTRODUCTION: Elhers-Danlos vascular syndrome type IV (EDS4) is a hereditary pathology of the connective tissue responsible for an increased risk of lethal arterial, uterine and digestive complications during and after pregnancy. PATIENTS AND METHODS: We describe the obstetrical care, the nature and frequency of complications related to pregnancy of patients with EDS4 and their relatives. RESULTS: Twenty-seven pregnancies were studied including 23 deliveries, 18 vaginal deliveries and five caesarean, no maternal death and two major life-threatening complications (8.7%) were recorded which could be directly linked to EDS4 (rupture of the biscupid valve pillar after vaginal delivery and a rupture of the caecum after a prophylactic caesarean). Ten deliveries underwent epidural anesthesia without complication. Six perineal injuries (33.3%) were observed. CONCLUSION: Pregnancy in patient with EDS4 needs obstetrical cares in a special unit's motivated medical team with intensive care and surgical disponibilities.


Subject(s)
Ehlers-Danlos Syndrome/complications , Ehlers-Danlos Syndrome/therapy , Pregnancy Complications/therapy , Pregnancy Outcome , Adult , Cecal Diseases/etiology , Cesarean Section , Delivery, Obstetric , Female , Heart Valve Diseases/etiology , Humans , Mitral Valve , Perineum/injuries , Pregnancy , Rupture, Spontaneous
13.
J Thromb Haemost ; 8(7): 1447-56, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20345718

ABSTRACT

SUMMARY: Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder characterized by severe and recurrent nosebleeds, mucocutaneous telangiectases, and, in some cases, life-threatening visceral arteriovenous malformations of various types, including pulmonary, hepatic, cerebral, and spinal. Gastrointestinal telangiectases are frequent and may cause severe bleeding. HHT type 1 results from mutations in ENG on chromosome 9 (coding for endoglin), and HHT type 2 results from mutations in ACVRL1 on chromosome 12 (coding for activin receptor-like kinase 1). Mutations of either of these two genes account for most clinical cases. In addition, mutations in MADH4 (encoding SMAD4), which cause a juvenile polyposis/HHT overlap syndrome, have been described, and recently, an HHT3 locus on chromosome 5 (5q31.3-5q32) has been reported. The mutated genes in HHT encode proteins that modulate transforming growth factor-beta superfamily signaling in vascular endothelial cells. Management of patients has changed considerably in the last 20 years, in terms of both treatment and the prevention of complications. The goal of this review was to describe the underlying molecular and cellular physiopathology, explore clinical and genetic diagnostic strategies for HHT, and present clinical management recommendations in order to treat symptomatic disease and to screen for vascular malformations.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic , Animals , Arteriovenous Malformations , Epistaxis , Humans , Signal Transduction/genetics , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/etiology , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/therapy
14.
Nature ; 463(7281): 671-5, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20130649

ABSTRACT

Obesity has become a major worldwide challenge to public health, owing to an interaction between the Western 'obesogenic' environment and a strong genetic contribution. Recent extensive genome-wide association studies (GWASs) have identified numerous single nucleotide polymorphisms associated with obesity, but these loci together account for only a small fraction of the known heritable component. Thus, the 'common disease, common variant' hypothesis is increasingly coming under challenge. Here we report a highly penetrant form of obesity, initially observed in 31 subjects who were heterozygous for deletions of at least 593 kilobases at 16p11.2 and whose ascertainment included cognitive deficits. Nineteen similar deletions were identified from GWAS data in 16,053 individuals from eight European cohorts. These deletions were absent from healthy non-obese controls and accounted for 0.7% of our morbid obesity cases (body mass index (BMI) >or= 40 kg m(-2) or BMI standard deviation score >or= 4; P = 6.4 x 10(-8), odds ratio 43.0), demonstrating the potential importance in common disease of rare variants with strong effects. This highlights a promising strategy for identifying missing heritability in obesity and other complex traits: cohorts with extreme phenotypes are likely to be enriched for rare variants, thereby improving power for their discovery. Subsequent analysis of the loci so identified may well reveal additional rare variants that further contribute to the missing heritability, as recently reported for SIM1 (ref. 3). The most productive approach may therefore be to combine the 'power of the extreme' in small, well-phenotyped cohorts, with targeted follow-up in case-control and population cohorts.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Obesity/genetics , Obesity/physiopathology , Penetrance , Adolescent , Adult , Age of Onset , Aging , Body Mass Index , Case-Control Studies , Child , Cognition Disorders/complications , Cognition Disorders/genetics , Cohort Studies , Europe , Female , Genome-Wide Association Study , Heterozygote , Humans , Inheritance Patterns/genetics , Male , Mutation/genetics , Obesity/complications , Reproducibility of Results , Sex Characteristics , Young Adult
16.
Hum Mol Genet ; 10(17): 1767-73, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11532986

ABSTRACT

Human malignant infantile osteopetrosis (arOP; MIM 259700) is a genetically heterogeneous autosomal recessive disorder of bone metabolism, which, if untreated, has a fatal outcome. Our group, as well as others, have recently identified mutations in the ATP6i (TCIRG1) gene, encoding the a3 subunit of the vacuolar proton pump, which mediates the acidification of the bone/osteoclast interface, are responsible for a subset of this condition. By sequencing the ATP6i gene in arOP patients from 44 unrelated families with a worldwide distribution we have now established that ATP6i mutations are responsible for approximately 50% of patients affected by this disease. The vast majority of these mutations (40 out of 42 alleles, including seven deletions, two insertions, 10 nonsense substitutions and 21 mutations in splice sites) are predicted to cause severe abnormalities in the protein product and are likely to represent null alleles. In addition, we have also analysed nine unrelated arOP patients from Costa Rica, where this disease is apparently much more frequent than elsewhere. All nine Costa Rican patients bore either or both of two missense mutations (G405R and R444L) in amino acid residues which are evolutionarily conserved from yeast to humans. The identification of ATP6i gene mutations in two families allowed us for the first time to perform prenatal diagnosis: both fetuses were predicted not to be affected and two healthy babies were born. This study contributes to the determination of genetic heterogeneity of arOP and allows further delineation of the other genetic defects causing this severe condition.


Subject(s)
Mutation , Osteopetrosis/genetics , Vacuolar Proton-Translocating ATPases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chloride Channels/genetics , Chromosomes, Human, Pair 11 , DNA Mutational Analysis , Exons , Female , Genes, Recessive , Haplotypes , Humans , Infant , Infant, Newborn , Introns , Male , Molecular Sequence Data , Osteopetrosis/enzymology , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Vacuoles/enzymology , Vacuoles/genetics
18.
Nat Genet ; 27(3): 277-85, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11242109

ABSTRACT

The molecular basis of X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) has remained elusive. Here we report hypomorphic mutations in the gene IKBKG in 12 males with EDA-ID from 8 kindreds, and 2 patients with a related and hitherto unrecognized syndrome of EDA-ID with osteopetrosis and lymphoedema (OL-EDA-ID). Mutations in the coding region of IKBKG are associated with EDA-ID, and stop codon mutations, with OL-EDA-ID. IKBKG encodes NEMO, the regulatory subunit of the IKK (IkappaB kinase) complex, which is essential for NF-kappaB signaling. Germline loss-of-function mutations in IKBKG are lethal in male fetuses. We show that IKBKG mutations causing OL-EDA-ID and EDA-ID impair but do not abolish NF-kappaB signaling. We also show that the ectodysplasin receptor, DL, triggers NF-kappaB through the NEMO protein, indicating that EDA results from impaired NF-kappaB signaling. Finally, we show that abnormal immunity in OL-EDA-ID patients results from impaired cell responses to lipopolysaccharide, interleukin (IL)-1beta, IL-18, TNFalpha and CD154. We thus report for the first time that impaired but not abolished NF-kappaB signaling in humans results in two related syndromes that associate specific developmental and immunological defects.


Subject(s)
Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Adolescent , Child , Child, Preschool , Codon, Terminator/genetics , Ectodermal Dysplasia/metabolism , Ectodysplasins , Genetic Linkage , Humans , I-kappa B Kinase , Immunity, Cellular , Immunologic Deficiency Syndromes/metabolism , Infant , Male , Membrane Proteins/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Syndrome , X Chromosome/genetics
19.
J Pediatr ; 138(1): 129-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11148528

ABSTRACT

Fanconi's anemia can be associated with growth retardation. We describe biologic growth hormone deficiency, isolated or associated with thyrotropin abnormality, and pituitary stalk interruption syndrome on magnetic resonance imaging of 5 patients with Fanconi's anemia. Growth hormone treatment produced catch-up growth in all cases. These findings suggest a common genetic origin.


Subject(s)
Fanconi Anemia/complications , Growth Disorders/etiology , Human Growth Hormone/deficiency , Pituitary Gland/abnormalities , Body Height/drug effects , Body Weight/drug effects , Child , Child, Preschool , Fanconi Anemia/blood , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Female , Follicle Stimulating Hormone/blood , Growth Disorders/blood , Growth Disorders/diagnosis , Growth Disorders/drug therapy , Human Growth Hormone/pharmacology , Human Growth Hormone/therapeutic use , Humans , Hydrocortisone/blood , Infant , Infant, Newborn , Luteinizing Hormone/blood , Magnetic Resonance Imaging , Male , Prolactin/blood , Retrospective Studies , Thyrotropin/blood , Thyroxine/blood , Treatment Outcome
20.
Bull Cancer ; 84(3): 264-72, 1997 Mar.
Article in French | MEDLINE | ID: mdl-9207872

ABSTRACT

Craniospinal irradiation is the gold standard treatment used in non metastatic medulloblastoma as prophylaxis against central nervous system (CNS) metastases. However, given the severe late effects caused by this procedure in children under 3 years of age, most pediatric oncologists are currently treating these patients with conventional chemotherapy in order to postpone or even avoid irradiation. In the French Society of Pediatric Oncology (SFOP) this attitude has been adopted since 1990. Among the patients treated without radiotherapy, 20 relapsed while on conventional chemotherapy and were entered in a study of high-dose chemotherapy (HDC) followed by autologous bone marrow transplantation (ABMT). Their median age at diagnosis was 23 months (range: 5-71 months) and the relapse occurred at a median time of 6.3 months after the initiation of chemotherapy. Complete surgical removal of the local relapse was the first treatment in 4/20 patients who were not evaluable for response. Sixteen of the 20 patients had measurable disease at the primary site (9 patients), or at metastatic sites (3 patients) or both (4 patients). The conditioning regimen consisted of combination busulfan 600 mg/m2 over 4 days and thiotepa 900 mg/m2 over 3 days. After recovery from aplasia, patients with a local relapse received local radiotherapy limited to posterior fossa. Among the 16 patients with measurable disease, 6 complete responses, 6 partial responses, 3 non response, were observed following HDC (response rate 75%). One patient was not evaluable. For the 20 patients, the event free survival (EFS) is 50%. Among the surviving patients, the median follow-up is 39.5 months post BMT (range: 21-92 months). Ten patients who developed a local relapse or local progression are alive with non evidence of disease (NED) without craniospinal irradiation. Among the 7 patients who developed a metastases or progression of metastases, only 1 is alive. Toxicity was high but manageable. One complication-related death occurred 1 month post BMT. With a 75% response rate, this HDC proved to be very efficient in relapsed medulloblastoma. A longer follow-up is necessary to demonstrate whether, after a local relapse, HDC could replace craniospinal irradiation as prophylaxis against CNS metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Bone Marrow Transplantation , Busulfan/administration & dosage , Busulfan/adverse effects , Cerebellar Neoplasms/mortality , Child , Child, Preschool , Combined Modality Therapy , Drug Administration Schedule , Female , Humans , Infant , Male , Medulloblastoma/mortality , Neoplasm Recurrence, Local/mortality , Survival Rate , Thiotepa/administration & dosage , Thiotepa/adverse effects , Transplantation, Autologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...