Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
J Clin Endocrinol Metab ; 101(5): 2099-104, 2016 05.
Article in English | MEDLINE | ID: mdl-27003306

ABSTRACT

CONTEXT: Premature ovarian insufficiency (POI) may be secondary to chemotherapy, radiotherapy, or environmental factors. Genetic causes are identified in 20-25% of cases, but most POI cases remain idiopathic. OBJECTIVE: This study aimed to identify new genes involved in POI and to characterize the implication of CPEB1 gene in POI. DESIGN AND SETTING: This was a case report and cohort study replicate conducted in academic medical centers. PATIENTS AND METHODS: A deletion including CPEB1 gene was first identified in a patient with primary amenorrhea. Secondly, 191 sporadic POI cases and 68 familial POI cases were included. For each patient, karyotype was normal and FMR1 premutation was excluded. Search for CPEB1 deletions was performed by quantitative multiplex PCR of short fluorescent fragments or DNA microarray analysis. Gene sequencing of CPEB1 was performed for 95 patients. RESULTS: We identified three patients carrying a microdeletion in band 15q25.2. The proximal breakpoint, for the three patients, falls within a low-copy repeat region disrupting the CPEB1 gene, which represents a strong candidate gene for POI as it is known to be implicated in oocyte meiosis. No mutation was identified by sequencing CPEB1 gene. Therefore, heterozygous deletion of CPEB1 gene leading to haploinsufficiency could be responsible for POI in humans. CONCLUSION: Microdeletions of CPEB1 were identified in 1.3% of patients with POI, whereas no mutation was identified. This microdeletion is rare but recurrent as it is mediated by nonallelic homologous recombination due to the existence of low-copy repeats in the region. This result demonstrates the importance of DNA microarray analysis in etiological evaluation and counseling of patients with POI.


Subject(s)
Gene Deletion , Menopause, Premature/genetics , Primary Ovarian Insufficiency/genetics , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Adult , Cohort Studies , Female , Humans , Mutation
2.
Med Leg J ; 84(2): 105-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26857073

ABSTRACT

Decapitation is the separation of the head from the neck. Accidental decapitation is rare, and very few cases are cited in the literature. In this case, the victim was asleep during an overnight trip with her head sticking out of the window, and she was decapitated by a truck travelling in the opposite direction. Lack of security grilles on windows, high-speed driving, narrow roads and night travel were contributing factors. This case is presented for its rarity and pattern of injuries during the fatal mishap and to consider possible preventive measures.


Subject(s)
Accidents/mortality , Decapitation/pathology , Crush Injuries/pathology , Decapitation/mortality , Female , Forensic Medicine/methods , Humans , India , Protective Devices/standards , Travel , Young Adult
3.
Ann Endocrinol (Paris) ; 71(3): 149-57, 2010 May.
Article in English | MEDLINE | ID: mdl-20362962

ABSTRACT

The Kallmann syndrome (KS) combines hypogonadotropic hypogonadism (HH) with anosmia. This is a clinically and genetically heterogeneous disease. KAL1, encoding the extracellular glycoprotein anosmin-1, is responsible for the X chromosome-linked recessive form of the disease (KAL1). Mutations in FGFR1 or FGF8, encoding fibroblast growth factor receptor-1 and fibroblast growth factor-8, respectively, underlie an autosomal dominant form with incomplete penetrance (KAL2). Mutations in PROKR2 and PROK2, encoding prokineticin receptor-2 and prokineticin-2, have been found in heterozygous, homozygous, and compound heterozygous states. These two genes are likely to be involved both in autosomal recessive monogenic (KAL3) and digenic/oligogenic KS transmission modes. Mutations in any of the above-mentioned KS genes have been found in less than 30% of the KS patients, which indicates that other genes involved in the disease remain to be discovered. Notably, KS may also be part of pleiotropic developmental diseases including CHARGE syndrome; this disease results in most cases from neomutations in CHD7 that encodes a chromodomain helicase DNA-binding protein.


Subject(s)
Extracellular Matrix Proteins/genetics , Kallmann Syndrome/genetics , Nerve Tissue Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Abnormalities, Multiple/genetics , Estrogen Replacement Therapy/methods , Female , Germ-Line Mutation , Humans , Hypogonadism/drug therapy , Hypogonadism/genetics , Hypogonadism/physiopathology , Kallmann Syndrome/physiopathology , Mutation , Mutation, Missense
4.
Sex Dev ; 2(4-5): 181-93, 2008.
Article in English | MEDLINE | ID: mdl-18987492

ABSTRACT

Kallmann syndrome (KS) combines hypogonadotropic hypogonadism and anosmia. Anosmia is related to the absence or hypoplasia of the olfactory bulbs and tracts. Hypogonadism is due to gonadotropin-releasing hormone (GnRH) deficiency, which presumably results from a failure of the embryonic migration of neuroendocrine GnRH cells from the olfactory epithelium to the forebrain. This failure could be a consequence of the early degeneration of olfactory nerve and terminal nerve fibres, because the latter normally act as guiding cues for the migration of GnRH cells. Defects in GnRH cell fate specification, differentiation, axon elongation or axon targeting to the hypothalamus median eminence may, however, also contribute to GnRH deficiency, at least in some genetic forms of the disease. To date, five KS genes have been identified, namely, FGFR1, FGF8, PROKR2, PROK2, and KAL1. Mutations in these genes, however, account for barely 30% of all KS cases. Mutations in FGFR1, encoding fibroblast growth factor receptor 1, underlie an autosomal dominant form of the disease. Mutations in PROKR2 and PROK2, encoding prokineticin receptor-2 and prokineticin-2, have been found in heterozygous, homozygous or compound heterozygous states. These two genes are likely to be involved both in monogenic recessive and digenic or oligogenic KS transmission modes. Finally, KAL1, encoding the extracellular glycoprotein anosmin-1, is responsible for the X chromosome-linked form of the disease. It is believed that anosmin-1 acts as an enhancer of FGF signalling and perhaps of prokineticin signalling too.


Subject(s)
Genetic Predisposition to Disease/genetics , Kallmann Syndrome/genetics , Kallmann Syndrome/pathology , Extracellular Matrix Proteins/genetics , Fibroblast Growth Factor 8/genetics , Gastrointestinal Hormones/genetics , Genotype , Humans , Male , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics
5.
Clin Exp Rheumatol ; 25(4 Suppl 45): S93-5, 2007.
Article in English | MEDLINE | ID: mdl-17949559

ABSTRACT

OBJECTIVE: Familial Mediterranean Fever (FMF) and TNF-Receptor Associated Periodic Syndrome (TRAPS) are two inheritable inflammatory disorders. They share some clinical manifestations but their treatments are different. We present here the case of an overlap syndrome of FMF and TRAPS in a patient carrying a mutation in both the MEFV and TNFRSF1A genes. CASE REPORT: A 20-year-old woman of Mediterranean origin had suffered since childhood from attacks of fever and arthritis, with skin and ophthalmic manifestations. The initial diagnosis was FMF. The symptoms responded poorly to colchicine but regressed with steroids. Genetic analysis revealed a homozygous M694V mutation in MEFV and a heterozygous R92Q mutation in TNFRSF1A. We discuss the complexity of this combined FMF-TRAPS phenotype. CONCLUSION: This case shows that mutations in MEFV and TNFRSF1A can occur together in a single patient, a condition that may modify its response to treatment. It would be interesting to evaluate the role of the R92Q mutation in TNFRSF1A in patients of Mediterranean origin with FMF unresponsive to colchicine.


Subject(s)
Colchicine/pharmacology , Cytoskeletal Proteins/genetics , Familial Mediterranean Fever , Gout Suppressants/pharmacology , Polymorphism, Single Nucleotide/genetics , Adult , Familial Mediterranean Fever/complications , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/genetics , Female , Humans , Mutation , Phenotype , Pyrin , Receptors, Tumor Necrosis Factor, Type I/genetics , Syndrome , TNF Receptor-Associated Factor 2 , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics
7.
Br J Dermatol ; 154(6): 1190-3, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16704654

ABSTRACT

A young woman patient had early and extensive familial Mediterranean fever (FMF)-related amyloidosis and pseudoxanthoma elasticum (PXE). She had the novel G1042S mutation in the ATP-binding cassette subfamily C member 6 (ABCC6) gene, responsible for PXE, and the mutation M694I in MEFV, the FMF gene. Both mutations were homozygous, in agreement with consanguinity in the parents. ABCC6 deficiency may have increased the severity of amyloidosis by increasing the deposition in target tissues of heparan sulphate, which colocalizes spatially and temporally with amyloid proteins, and/or by decreasing the therapeutic activity of colchicine.


Subject(s)
Amyloidosis/etiology , Familial Mediterranean Fever/complications , Pseudoxanthoma Elasticum/complications , ATP-Binding Cassette Transporters/genetics , Adult , Amino Acid Sequence , Amyloidosis/genetics , Amyloidosis/pathology , Familial Mediterranean Fever/genetics , Female , Humans , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/genetics , Mutation , Pseudoxanthoma Elasticum/genetics , Pseudoxanthoma Elasticum/pathology , Sequence Alignment
8.
Ann Rheum Dis ; 65(9): 1158-62, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16569687

ABSTRACT

OBJECTIVE: Tumour necrosis factor receptor-associated periodic syndrome (TRAPS) has been associated with several mutations in the TNF receptor super family 1A (TNFRSF1A), including most cysteine substitutions. However, the nature of two substitutions, P46L and R92Q, remains a topic of discussion. The aim of this study was to assess the actual role of these two sequence variations in a series of patients with TRAPS. METHODS: The main clinical data of 89 patients with TRAPS have been prospectively registered on a standard form. 84 patients or members of families with recurrent episodes of inflammatory symptoms spanning a period of more than 6 months and harbouring a TNFRSF1A mutation were studied. Clinical data have been analysed according to the nature of the mutation-P46L, R92Q or others. RESULTS: P46L is often seen in patients from Maghreb and is associated with a mild phenotype. P46L appears as a polymorphism with a non-specific role in inflammation. R92Q is associated with a variable phenotype and presents as a low-penetrance mutation. Interpreting these results will require a comparison with clinical signs and genetic background.


Subject(s)
Familial Mediterranean Fever/genetics , Mutation , Receptors, Tumor Necrosis Factor, Type I/genetics , Adolescent , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colchicine/therapeutic use , Familial Mediterranean Fever/blood , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/ethnology , Female , Genotype , Humans , Male , Phenotype , Prospective Studies , Receptors, Tumor Necrosis Factor/blood , Treatment Outcome , Tumor Necrosis Factor-alpha/analysis
9.
Rev Med Interne ; 24(12): 781-5, 2003 Dec.
Article in French | MEDLINE | ID: mdl-14656637

ABSTRACT

PURPOSE: Tumor necrosis factor receptor superfamily 1A associated periodic syndrome (TRAPS) belongs to the group of hereditary fever syndromes, also called hereditary auto-inflammatory syndromes. CURRENT KNOWLEDGE AND KEY POINTS: The diagnosis of TRAPS should be evoked in presence of the following clinical signs, whatever the population of the affected patients. TRAPS acute inflammatory access, of 1 to 3 weeks' duration, is characterised by the presence of fever, abdominal pain, myalgias, various types of skin rash including erysepela-like erythema. Long term inflammatory response can lead to AA amyloidosis. Genetic testing will confirm the diagnosis when showing a mutation in the extracellular part of the TNFRSF1A receptor. Therapeutic management of TRAPS is not definitely established. Daily colchicine does not seem to prevent efficiently inflammatory attacks. Corticosteroids, in contrast can attenuate the intensity and diminish the duration of attacks. FUTURE PROSPECTS AND PROJECTS: The value of biological agents that inhibits TNF action is not yet completely determined in TRAPS. Mechanisms of the disease are not yet elucidated. In some families with specific mutations, a relative soluble TNF receptor deficiency has been found in the plasma. However this mechanism does not account for what is observed in other kindreds.


Subject(s)
Antigens, CD/genetics , Antigens, CD/physiology , Familial Mediterranean Fever/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Abdominal Pain/etiology , Amyloid/metabolism , Apoptosis , Colchicine/therapeutic use , Diagnosis, Differential , Erythema/etiology , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/physiopathology , Gout Suppressants/therapeutic use , Humans , Inflammation , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Am J Med Genet A ; 119A(3): 279-82, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12784292

ABSTRACT

Huntington disease (HD) is a neurodegenerative disorder caused by the abnormal expansion of CAG repeats in the HD gene on chromosome 4p16.3. Past studies have shown that the size of expanded CAG repeat is inversely associated with age at onset (AO) of HD. It is not known whether the normal Huntington allele size influences the relation between the expanded repeat and AO of HD. Data collected from two independent cohorts were used to test the hypothesis that the unexpanded CAG repeat interacts with the expanded CAG repeat to influence AO of HD. In the New England Huntington Disease Center Without Walls (NEHD) cohort of 221 HD affected persons and in the HD-MAPS cohort of 533 HD affected persons, we found evidence supporting an interaction between the expanded and unexpanded CAG repeat sizes which influences AO of HD (P = 0.08 and 0.07, respectively). The association was statistically significant when both cohorts were combined (P = 0.012). The estimated heritability of the AO residual was 0.56 after adjustment for normal and expanded repeats and their interaction. An analysis of tertiles of repeats sizes revealed that the effect of the normal allele is seen among persons with large HD repeat sizes (47-83). These findings suggest that an increase in the size of the normal repeat may mitigate the expression of the disease among HD affected persons with large expanded CAG repeats.


Subject(s)
Huntington Disease/genetics , Trinucleotide Repeats , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Child , Child, Preschool , Cohort Studies , Humans , Middle Aged , New England , Probability , Survival Rate
11.
Dermatology ; 206(3): 257-9, 2003.
Article in English | MEDLINE | ID: mdl-12673085

ABSTRACT

The Muckle-Wells syndrome is a rare autosomal dominant disorder belonging to the group of hereditary fever syndromes. The chronic infantile neurological cutaneous and articular (CINCA) syndrome is a systemic inflammatory disorder of unknown etiology with neonatal onset. They are considered as two different entities. We report the case of a 36-year-old man suffering since birth from a nonpruritic generalized urticaria, with inflammatory flares, joint manifestations and progressive deafness requiring a bilateral hearing aid. An initial diagnosis of Muckle-Wells syndrome was made. However, the patient had an unusual clinical presentation with slightly dysmorphic facial appearance, clubbing of the fingers, mild mental retardation and papilledema. After a genetic advice, a diagnosis of CINCA syndrome was made. Search for mutations in the CIAS1 gene revealed a new mutation in a heterozygous state. This case report really raises the question of a link between these two inflammatory diseases. Further studies are needed to confirm the involvement of mutations of the CIAS1 gene in CINCA syndrome.


Subject(s)
Arthritis/genetics , Blood Proteins/genetics , Carrier Proteins/genetics , Deafness/genetics , Face/abnormalities , Urticaria/genetics , Adult , Chronic Disease , Diagnosis, Differential , Fingers/abnormalities , Humans , Male , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein , Syndrome
12.
Neurology ; 58(6): 965-7, 2002 Mar 26.
Article in English | MEDLINE | ID: mdl-11914418

ABSTRACT

The authors report a large series of patients with Huntington disease (HD)-like phenotype without CAG repeat expansions in the IT15 gene that were screened for the newly identified CAG/CTG expansion in the gene encoding junctophilin-3. Normal alleles in controls had from 8 to 28 repeats. A single patient of North African origin with typical HD carried an allele with 50 uninterrupted repeats, representing approximately 2% of the non-IT15 HD patients tested. Therefore, further genetic heterogeneity is expected in HD.


Subject(s)
Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Adolescent , Adult , Aged , Alleles , Brain/pathology , Child , Child, Preschool , Female , Humans , Huntington Disease/pathology , Infant , Male , Membrane Proteins/genetics , Middle Aged
13.
Isr Med Assoc J ; 3(11): 803-4, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11729572

ABSTRACT

BACKGROUND: Familial Mediterranean fever is a genetic disease in which some characteristic gene mutations have been found. OBJECTIVES: To analyze the phenotype-genotype correlations in North African Jews and Armenians with FMF. METHODS: We studied MEFV gene mutations and phenotype-genotype correlations in North African Jews and Armenians with Familial Mediterranean Fever living in France. RESULTS: M694V mutation was the most common mutation in Jews and in Armenians. Patients with M680I homozygosity or M680I/M694V compound heterozygosity had a phenotype as severe as patients with M694V homozygosity. CONCLUSIONS: This study characterizes the phenotype-genotype in specific ethnic groups of patients with FMF.


Subject(s)
Familial Mediterranean Fever/genetics , Mutation/genetics , Proteins/genetics , White People/genetics , Africa, Northern/ethnology , Armenia/ethnology , Cytoskeletal Proteins , DNA Mutational Analysis , Electrophoresis, Agar Gel , Exons/genetics , France , Genotype , Humans , Jews/genetics , Phenotype , Pyrin , Severity of Illness Index
14.
Kidney Int ; 59(5): 1677-82, 2001 May.
Article in English | MEDLINE | ID: mdl-11318938

ABSTRACT

BACKGROUND: The recent identification of genes responsible for syndromes of periodic fever with amyloidosis has opened the way to a molecular diagnosis of hereditary AA amyloidosis. METHODS: A Belgian woman presented for genetic counseling. Three first-degree relatives had a diagnosis of renal amyloidosis with a history of recurrent fever and inflammatory episodes. Medical records and pathological specimens were obtained from all physicians who had been in charge of her three relatives. Immunohistochemical staining was performed on paraffin-embedded material. A mutation search was performed in the MEFV (Mediterranean fever) and tumor necrosis factor receptor 1 (TNFR1 or TNFRSF1A) genes causing familial Mediterranean fever (FMF) and tumor necrosis factor receptor-associated periodic syndrome (TRAPS), respectively. RESULTS: The family history was consistent with autosomal-dominant transmission of periodic fever with arthralgias, abdominal pain, and eventual AA amyloidosis involving the kidneys, digestive tract, and thyroid. Recurrent amyloidosis in kidney graft was demonstrated in one patient and was suspected in the other. A novel heterozygous mutation (C55S) in TNFRSF1A was identified in the affected patient available for genetic testing but not in the asymptomatic woman requiring counseling. No mutation was detected in MEFV. CONCLUSIONS: We report a novel mutation (C55S) in TNFRSF1A, resulting in autosomal-dominant periodic fever and AA amyloidosis. This condition, known as TRAPS, should be added to the differential diagnosis of hereditary renal amyloidosis, with obvious implications for management and genetic counseling.


Subject(s)
Amyloidosis/complications , Amyloidosis/genetics , Antigens, CD/genetics , Familial Mediterranean Fever/complications , Familial Mediterranean Fever/genetics , Mutation , Receptors, Tumor Necrosis Factor/genetics , Adult , Aged , Amyloidosis/pathology , Cytoskeletal Proteins , DNA Mutational Analysis , Female , Genes, Dominant , Humans , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Pedigree , Proteins/genetics , Pyrin , Receptors, Tumor Necrosis Factor, Type I
16.
Eur J Hum Genet ; 9(1): 51-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11175300

ABSTRACT

Seventy-nine unrelated Lebanese patients were tested for 15 mutations in the MEFV gene: A761H, A744S, V726A, K695R, M694V, M694I, M694del, M6801 (G --> C), M680I (G --> A) in exon 10, F479L in exon 5, P369S in exon 3, T267I, E167D and E148Q in exon 2, using PCR digestion, ARMS, DGGE and/or sequencing. Mutations were detected in patients belonging to all communities, most interestingly the Maronite, Greek orthodox, Greek catholic, Syriac and Chiite communities. The most frequent mutations are M694V and V726A (27% and 20% of the total alleles respectively). M694I, E148Q and M680I mutations account respectively for 9%, 8% and 5%. Each of the K695R, E167D and F479L mutations was observed once and all the remaining mutations were not encountered. Of the alleles 33% do not carry any of the studied mutations. The mutation spectra, clinical features and severity of the disease differed among the Lebanese communities. The genotype-phenotype analysis showed a significant association (P < 0.001) between amyloidosis and the presence of mutations at codon 694 in exon 10 (both M694V and M694I). None of the patients carrying other mutations developed amyloidosis.


Subject(s)
Familial Mediterranean Fever/genetics , Proteins/genetics , Amyloidosis/genetics , Amyloidosis/pathology , Cytoskeletal Proteins , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Familial Mediterranean Fever/pathology , Gene Frequency , Genotype , Humans , Lebanon , Mutation , Pyrin , Religion , Severity of Illness Index
17.
Eur J Intern Med ; 11(5): 242-244, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11025247
18.
Arthritis Rheum ; 43(7): 1535-42, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10902757

ABSTRACT

OBJECTIVE: To characterize both phenotypic (clinical features and magnetic resonance imaging [MRI] findings) and genotypic aspects of autosomal-dominant recurrent fever, also known as tumor necrosis factor receptor (TNFR)-associated periodic syndrome (TRAPS), in a French family and to investigate the role of the mutated 55-kd tumor necrosis factor alpha (TNFalpha) receptor (TNFR1) in the pathogenesis of the disease. METHODS: The coding region of TNFR1 was sequenced in 2 individuals with TRAPS (the propositus and her grandfather) and in 3 clinically unaffected relatives. Expression of soluble TNFR1 (sTNFR1) was investigated in 3 of the family members carrying a C30S mutation in TNFR1, and was compared with the levels of soluble TNFR2 (sTNFR2) by enzyme-linked immunosorbent assay. The membrane TNFR1 expression was then compared with membrane TNFR2 levels at the surface of peripheral blood mononuclear cells by flow cytometric analysis. The clinical heterogeneity in this French family was investigated by searching polymorphic variants in the TNFalpha promoter by DNA sequencing. RESULTS: Both the disease course and the clinical presentation in the propositus were highly indicative of TRAPS. MRI study of the segmental inflammatory process in the limbs showed abnormal signals in the muscle and subcutaneous tissue without involvement of adjacent joints or fascia. A novel missense mutation, C30S, in the first extracellular N-terminal cysteine-rich domain (CRD1) of TNFR1 was characterized in the propositus, her affected grandfather, and her clinically unaffected father. Expression of membrane TNFR1 at the surface of monocytes and polymorphonuclear leukocytes, as well as the levels of sTNFR1 in serum when the disease was not active were not modified in the 3 individuals carrying the TNFR1 C30S mutation. In contrast, during attacks, sTNFR1 levels remained abnormally low, as compared with the levels in unrelated patients with active adult-onset systemic Still's disease. The clinical heterogeneity could not be explained by a polymorphic variant in the TNFalpha promoter. CONCLUSION: TRAPS is a distinct clinical and radiologic disease entity that is responsible for recurrent fever and migratory cellulitis-like processes with localized myositis. We have identified a novel TNFR1 mutation, C30S, that is located in the CRD1 domain in a French family affected by the disease. This mutation seems to affect the level of sTNFR1, which did not increase in the propositus during inflammatory attacks.


Subject(s)
Antigens, CD/genetics , Fever/genetics , Genes, Dominant , Mutation, Missense , Myositis/genetics , Receptors, Tumor Necrosis Factor/genetics , Adolescent , Adult , Antigens, CD/blood , DNA Primers/analysis , Enzyme-Linked Immunosorbent Assay , Female , Fever/blood , Flow Cytometry , France , Humans , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Myositis/blood , Myositis/diagnosis , Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recurrence , Sequence Analysis, DNA
19.
Am J Med Genet ; 92(4): 241-6, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10842288

ABSTRACT

Familial Mediterranean fever (FMF) is an autosomal recessively inherited disease affecting patients of the Mediterranean basin. FMF is characterized by recurrent episodes of fever accompanied with topical signs of inflammation. Some patients can develop a renal amyloidosis associated (AA) amyloidosis. The administration of colchicine is an effective preventive treatment of both the attacks and amyloidosis. The FMF gene (MEFV) was cloned and missense mutations were found to be responsible for the disease. We investigated a large series of 303 unselected and unrelated patients of various ethnic backgrounds with a clinical suspicion of FMF to confirm or invalidate the diagnosis of FMF and to determine the spectrum of MEFV mutations. Molecular analysis focused on all the most frequent mutations identified so far, and an exhaustive analysis of exon 10, containing the mutational hotspot, was performed through DNA sequencing. Sixty-two percent of Sephardic, North African Arabs, Armenian and Turkish patients were either homozygous or compound heterozygous for MEFV mutations. In other populations surrounding the Mediterranean Sea such as Greek, Italian, Portuguese, Kurdish and Lebanese populations, mutations were also found. In general, patients without Mediterranean origin had no mutations in the MEFV gene. Two new mis-sense mutations were identified in exon 10 of the MEFV gene: the S675N in an Italian patient and the M680L in a French patient without any known at-risk ethnic ancestry.


Subject(s)
Familial Mediterranean Fever/genetics , Proteins/genetics , Amino Acid Substitution , Base Sequence , Cytoskeletal Proteins , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Familial Mediterranean Fever/ethnology , Familial Mediterranean Fever/pathology , Genotype , Heterozygote , Homozygote , Humans , Mutation , Point Mutation , Pyrin
20.
QJM ; 93(4): 223-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10787450

ABSTRACT

The diagnosis of familial Mediterranean fever (FMF) has until recently been based on clinical signs alone. Discovery of the MEFV gene has enabled a molecular approach to diagnosis, which is already well established for diagnosing typical clinical forms of FMF. We evaluated the utility of this molecular approach in a large series of patients with various clinical presentations and ethnic origins. We looked for mutations in the MEFV gene in 303 unselected consecutive patients with a variable (from high to low) clinical suspicion of FMF. Two mutations were found in 133 patients (44%). In 22 patients (7%), the clinical diagnosis of FMF was unlikely according to the Tel Hashomer clinical criteria. Our results suggest that the spectrum of FMF-associated signs is broader than previously believed. Wider indications for genotyping should lead to more frequent diagnosis of FMF.


Subject(s)
Familial Mediterranean Fever/diagnosis , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Diagnosis, Differential , Familial Mediterranean Fever/ethnology , Familial Mediterranean Fever/genetics , Genotype , Humans , Infant , Middle Aged , Mutation/genetics , Pedigree , Predictive Value of Tests , Prospective Studies , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...