Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
BMC Med Genomics ; 17(1): 73, 2024 Mar 06.
Article En | MEDLINE | ID: mdl-38448973

About 5-10% of neurofibromatosis type 1 (NF1) patients exhibit large genomic germline deletions that remove the NF1 gene and its flanking regions. The most frequent NF1 large deletion is 1.4 Mb, resulting from homologous recombination between two low copy repeats. This "type-1" deletion is associated with a severe clinical phenotype in NF1 patients, with several phenotypic manifestations including learning disability, a much earlier development of cutaneous neurofibromas, an increased tumour risk, and cardiovascular malformations. NF1 adjacent co-deleted genes could act as modifier loci for the specific clinical manifestations observed in deleted NF1 patients. Furthermore, other genetic modifiers (such as CNVs) not located at the NF1 locus could also modulate the phenotype observed in patients with large deletions. In this study, we analysed 22 NF1 deletion patients by genome-wide array-CGH with the aim (1) to correlate deletion length to observed phenotypic features and their severity in NF1 deletion syndrome, and (2) to identify whether the deletion phenotype could also be modulated by copy number variations elsewhere in the genome. We then review the role of co-deleted genes in the 1.4 Mb interval of type-1 deletions, and their possible implication in the main clinical features observed in this high-risk group of NF1 patients.


DNA Copy Number Variations , Skin Neoplasms , Humans , Comparative Genomic Hybridization , Genomics , Phenotype
2.
J Mol Diagn ; 26(2): 150-157, 2024 Feb.
Article En | MEDLINE | ID: mdl-38008284

Neurofibromatosis type-1 is a genetic disorder caused by loss-of-function variants in the tumor-suppressor NF1. Approximately 4% to 11% of neurofibromatosis type-1 patients have a NF1 locus complete deletion resulting from nonallelic homologous recombination between low copy repeats. Codeleted genes probably account for the more severe phenotype observed in NF1-deleted patients. This genotype-phenotype correlation highlights the need for a detailed molecular description. A droplet digital PCR (ddPCR) set along the NF1 locus was designed to delimitate the three recurrent NF1 deletion breakpoints. The ddPCR was tested in 121 samples from nonrelated NF1-deleted patients. Classification based on ddPCR versus multiplex ligation-dependent probe amplification (MLPA) was compared. In addition, microsatellites were analyzed to identify parental origin of deletions. ddPCR identified 77 type-1 (64%), 20 type-2 (16%), 7 type-3 (6%), and 17 atypical deletions (14%). The results were comparable with MLPA, except for three atypical deletions misclassified as type-2 using MLPA, for which the SUZ12 gene was not deleted. A significant maternal bias (25 of 30) in the origin of deletions was identified. This study proposes a fast and efficient ddPCR quantification to allow fine NF1 deletion classification. It indicates that ddPCR can be implemented easily into routine diagnosis to complement the techniques dedicated to NF1 point variant identification. This new tool may help unravel the genetic basis conditioning phenotypic variability in NF1-deleted patients and offer tailored genetic counseling.


Neurofibromatosis 1 , Humans , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics , Multiplex Polymerase Chain Reaction , Homologous Recombination , Phenotype , Family , Gene Deletion
3.
Br J Dermatol ; 190(2): 226-243, 2024 Jan 23.
Article En | MEDLINE | ID: mdl-37831592

BACKGROUND: Neurofibromatosis type 1 (NF1) is characterized by the highly variable and unpredictable development of benign peripheral nerve sheath tumours: cutaneous (cNFs), subcutaneous (scNFs) and plexiform (pNFs) neurofibromas. OBJECTIVES: To identify neurofibroma modifier genes, in order to develop a database of patients with NF1. METHODS: All patients were phenotypically evaluated by a medical practitioner using a standardized questionnaire and the causal NF1 variant identified. We enrolled 1333 patients with NF1 who were genotyped for > 7 million common variants. RESULTS: A genome-wide association case-only study identified a significant association with 9q21.33 in the pNF phenotype in the discovery cohort. Twelve, three and four regions suggestive of association at the P ≤ 1 × 10-6 threshold were identified for pNFs, cNFs and scNFs, respectively. Evidence of replication was observed for 4, 2 and 6 loci, including 168 candidate modifier protein-coding genes. Among the candidate modifier genes, some were implicated in the RAS-mitogen-activated protein kinase pathway, cell-cycle control and myelination. Using an original CRISPR/Cas9-based functional assay, we confirmed GAS1 and SPRED2 as pNF and scNF candidate modifiers, as their inactivation specifically affected NF1-mutant Schwann cell growth. CONCLUSIONS: Our study may shed new light on the pathogenesis of NF1-associated neurofibromas and will, hopefully, contribute to the development of personalized care for patients with this deleterious and life-threatening condition.


Neurofibroma, Plexiform , Neurofibroma , Neurofibromatosis 1 , Humans , Neurofibromatosis 1/genetics , Neurofibroma, Plexiform/complications , Neurofibroma, Plexiform/genetics , Genome-Wide Association Study , Neurofibroma/complications , Neurofibroma/genetics , Genotype , Repressor Proteins/genetics
5.
Eur J Hum Genet ; 31(11): 1337-1341, 2023 11.
Article En | MEDLINE | ID: mdl-37670079

Amelogenesis imperfecta (AI) is a group of rare genetic conditions characterized by quantitative and/or qualitative tooth enamel alterations. AI can manifest as an isolated trait or as part of a syndrome. Recently, five biallelic disease-causing variants in the RELT gene were identified in 7 families with autosomal recessive amelogenesis imperfecta (ARAI). RELT encodes an orphan receptor in the tumor necrosis factor (TNFR) superfamily expressed during tooth development, with unknown function. Here, we report one Brazilian and two French families with ARAI and a distinctive hypomineralized phenotype with hypoplastic enamel, post-eruptive enamel loss, and occlusal attrition. Using Next Generation Sequencing (NGS), four novel RELT variants were identified (c.120+1G>A, p.(?); c.120+1G>T, p.(?); c.193T>C, p.(Cys65Arg) and c.1260_1263dup, p.(Arg422Glyfs*5)). Our findings extend the knowledge of ARAI dental phenotypes and expand the disease-causing variants spectrum of the RELT gene.


Amelogenesis Imperfecta , Humans , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/pathology , Receptors, Tumor Necrosis Factor/genetics , Phenotype , Brazil , Pedigree
7.
Hum Genet ; 142(1): 1-9, 2023 Jan.
Article En | MEDLINE | ID: mdl-35941319

Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but highly variable expressivity. In most patients, Next Generation Sequencing (NGS) technologies allow the identification of a loss-of-function pathogenic variant in the NF1 gene, a negative regulator of the RAS-MAPK pathway. We describe the 5-year diagnosis wandering of a patient with a clear NF1 clinical diagnosis, but no molecular diagnosis using standard molecular technologies. The patient presented with a typical NF1 phenotype but NF1 targeted NGS, NF1 transcript analysis, MLPA, and array comparative genomic hybridization failed to reveal a genetic aberration. After 5 years of unsuccessful investigations, trio WGS finally identified a de novo mosaic (VAF ~ 14%) 24.6 kb germline deletion encompassing the promoter and first exon of NF1. This case report illustrates the relevance of WGS to detect structural variants including copy number variants that would be missed by alternative approaches. The identification of the causal pathogenic variant allowed a tailored genetic counseling with a targeted non-invasive prenatal diagnosis by detecting the deletion in plasmatic cell-free DNA from the proband's pregnant partner. This report clearly highlights the need to make WGS a clinically accessible test, offering a tremendous opportunity to identify a molecular diagnosis for otherwise unsolved cases.


Neurofibromatosis 1 , Humans , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics , Genes, Neurofibromatosis 1 , Comparative Genomic Hybridization , Exons , Whole Genome Sequencing
8.
Hum Mutat ; 43(12): 2308-2323, 2022 12.
Article En | MEDLINE | ID: mdl-36273432

Modeling splicing is essential for tackling the challenge of variant interpretation as each nucleotide variation can be pathogenic by affecting pre-mRNA splicing via disruption/creation of splicing motifs such as 5'/3' splice sites, branch sites, or splicing regulatory elements. Unfortunately, most in silico tools focus on a specific type of splicing motif, which is why we developed the Splicing Prediction Pipeline (SPiP) to perform, in one single bioinformatic analysis based on a machine learning approach, a comprehensive assessment of the variant effect on different splicing motifs. We gathered a curated set of 4616 variants scattered all along the sequence of 227 genes, with their corresponding splicing studies. The Bayesian analysis provided us with the number of control variants, that is, variants without impact on splicing, to mimic the deluge of variants from high-throughput sequencing data. Results show that SPiP can deal with the diversity of splicing alterations, with 83.13% sensitivity and 99% specificity to detect spliceogenic variants. Overall performance as measured by area under the receiving operator curve was 0.986, better than SpliceAI and SQUIRLS (0.965 and 0.766) for the same data set. SPiP lends itself to a unique suite for comprehensive prediction of spliceogenicity in the genomic medicine era. SPiP is available at: https://sourceforge.net/projects/splicing-prediction-pipeline/.


RNA Splice Sites , RNA Splicing , Humans , Bayes Theorem , RNA Splicing/genetics , Exons/genetics , RNA Splice Sites/genetics , Machine Learning , Introns/genetics
9.
J Clin Endocrinol Metab ; 107(1): e224-e235, 2022 01 01.
Article En | MEDLINE | ID: mdl-34406383

CONTEXT: Enthesopathies are the determinant of a poor quality of life in adults with X-linked hypophosphatemia (XLH). OBJECTIVE: To describe the prevalence of patients with enthesopathies and to identify the risk factors of having enthesopathies. METHODS: Retrospective study in the French Reference Center for Rare Diseases of the Calcium and Phosphate Metabolism between June 2011 and December 2020. Adult XLH patients with full body X-rays performed using the EOS® low-dose radiation system and clinical data collected from medical records. The main outcome measures were demographics, PHEX mutation, conventional treatment, and dental disease with the presence of enthesopathies. RESULTS: Of the 114 patients included (68% women, mean age 42.2 ± 14.3 years), PHEX mutation was found in 105 patients (94.6%), 86 (77.5%) had been treated during childhood. Enthesopathies (spine and/or pelvis) were present in 67% of the patients (n = 76). Patients with enthesopathies were significantly older (P = .001) and more frequently reported dental disease collected from medical records (P = .03). There was no correlation between the PHEX mutations and the presence of enthesopathies. Sixty-two patients had a radiographic dental examination in a reference center. Severe dental disease (number of missing teeth, number of teeth endodontically treated, alveolar bone loss, and proportion of patients with 5 abscesses or more) was significantly higher in patients with enthesopathies. CONCLUSION: Adult XLH patients have a high prevalence of enthesopathies in symptomatic adults patients with XLH seen in a reference center. Age and severe dental disease were significantly associated with the presence of enthesopathies.


Enthesopathy/epidemiology , Familial Hypophosphatemic Rickets/physiopathology , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Quality of Life , Adult , Enthesopathy/genetics , Enthesopathy/pathology , Female , Follow-Up Studies , Humans , Male , Prevalence , Prognosis , Retrospective Studies , Risk Factors
10.
Eur J Hum Genet ; 30(3): 291-297, 2022 03.
Article En | MEDLINE | ID: mdl-34897289

Individuals with the three base pair deletion NM_000267.3(NF1):c.2970_2972del p.(Met992del) have been recognised to present with a milder neurofibromatosis type 1 (NF1) phenotype characterised by café-au-lait macules (CALs) and intertriginous freckling, as well as a lack of cutaneous, subcutaneous and plexiform neurofibromas and other NF1-associated complications. Examining large cohorts of patients over time with this specific genotype is important to confirm the presentation and associated risks of this variant across the lifespan. Forty-one individuals with the in-frame NF1 deletion p.Met992del were identified from 31 families. Clinicians completed a standardised clinical questionnaire for each patient and the resulting data were collated and compared to published cohorts. Thirteen patients have been previously reported, and updated clinical information has been obtained for these individuals. Both CALs and intertriginous freckling were present in the majority of individuals (26/41, 63%) and the only confirmed features in 11 (27%). 34/41 (83%) of the cohort met NIH diagnostic criteria. There was a notable absence of all NF1-associated tumour types (neurofibroma and glioma). Neurofibroma were observed in only one individual-a subcutaneous lesion (confirmed histologically). Nineteen individuals were described as having a learning disability (46%). This study confirms that individuals with p.Met992del display a mild tumoural phenotype compared to those with 'classical', clinically diagnosed NF1, and this appears to be the case longitudinally through time as well as at presentation. Learning difficulties, however, appear to affect a significant proportion of NF1 subjects with this phenotype. Knowledge of this genotype-phenotype association is fundamental to accurate prognostication for families and caregivers.


Neurofibroma , Neurofibromatosis 1 , Cafe-au-Lait Spots/genetics , Genetic Association Studies , Humans , Longitudinal Studies , Neurofibroma/genetics , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology
11.
J Clin Endocrinol Metab ; 107(4): e1367-e1373, 2022 03 24.
Article En | MEDLINE | ID: mdl-34897474

CONTEXT: Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disease caused by mutations in the tumor suppressor gene MEN1. The uncertainty of pathogenicity of MEN1 variants complexifies the selection of the patients likely to benefit from specific care. OBJECTIVE: MEN1-mutated patients should be offered tailored tumor screening and genetic counseling. We present a patient with hyperparathyroidism for whom genetic analysis identified a variant of uncertain significance in the MEN1 gene (NM_130799.2): c.654G > T p.(Arg218=). Additional functional genetic tests were performed to classify the variant as pathogenic and allowed prenatal testing. DESIGN: Targeted next generation sequencing identified a synonymous variant in the MEN1 gene in a 26-year-old male with symptomatic primary hyperparathyroidism. In silico and in vitro genetic tests were performed to assess variant pathogenicity. RESULTS: Genetic testing of the proband's unaffected parents showed the variant occurred de novo. Transcript study showed a splicing defect leading to an in-frame deletion. The classification of the MEN1 variant as pathogenic confirmed the diagnosis of MEN1 and recommended an adapted medical care and follow-up. Pathogenic classification also allowed to propose a genetic counseling to the proband and his wife. Noninvasive prenatal diagnosis was performed with a personalized medicine-based protocol by detection of the paternally inherited variant in maternal plasmatic cell free DNA, using digital PCR. CONCLUSION: We showed that functional genetic analysis can help to assess the pathogenicity of a MEN1 variant with crucial consequences for medical care and genetic counseling decisions.


Hyperparathyroidism , Multiple Endocrine Neoplasia Type 1 , Noninvasive Prenatal Testing , Adult , Female , Genetic Testing , Humans , Hyperparathyroidism/genetics , Male , Multiple Endocrine Neoplasia Type 1/genetics , Paternal Inheritance , Pregnancy
12.
Cancers (Basel) ; 13(12)2021 Jun 13.
Article En | MEDLINE | ID: mdl-34199217

Complete deletion of the NF1 gene is identified in 5-10% of patients with neurofibromatosis type 1 (NF1). Several studies have previously described particularly severe forms of the disease in NF1 patients with deletion of the NF1 locus, but comprehensive descriptions of large cohorts are still missing to fully characterize this contiguous gene syndrome. NF1-deleted patients were enrolled and phenotypically characterized with a standardized questionnaire between 2005 and 2020 from a large French NF1 cohort. Statistical analyses for main NF1-associated symptoms were performed versus an NF1 reference population. A deletion of the NF1 gene was detected in 4% (139/3479) of molecularly confirmed NF1 index cases. The median age of the group at clinical investigations was 21 years old. A comprehensive clinical assessment showed that 93% (116/126) of NF1-deleted patients fulfilled the NIH criteria for NF1. More than half had café-au-lait spots, skinfold freckling, Lisch nodules, neurofibromas, neurological abnormalities, and cognitive impairment or learning disabilities. Comparison with previously described "classic" NF1 cohorts showed a significantly higher proportion of symptomatic spinal neurofibromas, dysmorphism, learning disabilities, malignancies, and skeletal and cardiovascular abnormalities in the NF1-deleted group. We described the largest NF1-deleted cohort to date and clarified the more severe phenotype observed in these patients.

13.
Gene ; 753: 144793, 2020 Aug 30.
Article En | MEDLINE | ID: mdl-32446918

BACKGROUND: Fragile X syndrome (FXS) is a monogenic disorder and a common cause of intellectual disability (ID). Up to now, very few pathological variants other than the typical CGG-repeat expansion have been reported in the FMR1 gene. METHODS: A panel of 56 intellectual disability (ID) genes including the FMR1 gene was sequenced in a cohort of 300 patients with unexplained ID. To determine the effect of a new FMR1 variant, total RNA from peripheral blood cells was reverse transcribed, amplified by polymerase chain reaction and sequenced. RESULTS: We report a novel G to A point variant (c.801G > A) located at the last nucleotide of exon 8 in the FMR1 gene in one patient with ID. Direct sequencing of the RT-PCR products revealed that the transcript from the allele with G to A variant skips exon 8 entirely, resulting in a joining of exons 7 and 9. Skipping of exon 8 may result in an abnormal FMR1 protein (FMRP), which removes the highly conserved region that encoding the KH1 domain of FMRP. CONCLUSIONS: This report describes for the first time that a synonymous variant in the FMR1 gene is associated with an error in mRNA processing, leading preferentially to the production of an aberrant transcript without exon 8. This splice variant was associated with an unspecific clinical presentation, suggesting the need for more detailed investigation of silent variants in ID patients with a large spectrum of phenotypes.


Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Intellectual Disability/genetics , Adolescent , Adult , Alleles , Cohort Studies , Exons , Humans , Male , Mutation , Phenotype , Polymorphism, Single Nucleotide , Protein Domains , RNA Splicing , Sequence Analysis, RNA/methods , Silent Mutation , Trinucleotide Repeat Expansion
14.
Front Med (Lausanne) ; 7: 77, 2020.
Article En | MEDLINE | ID: mdl-32211415

Two patients presented simultaneously to our hospital with distinct clinical features associated with the presence of anti-MDA5 antibodies: the first one was admitted for a skin rash resembling to a toxic epidermal necrosis (Lyell syndrome) and the second one presented with pulmonary manifestations attributed to a diffuse fibrosing interstitial pneumonitis on chest CT-scan. In addition to the skin lesions involving 40% of the body surface area, the first patient developed a rapid diffuse interstitial pneumonitis with respiratory distress justifying the initiation of a systemic immunosuppressive treatment. However, she died 3 weeks after her admission from mesenteric thrombosis associated with septic shock. The second patient respiratory condition worsened despite an intensive immunosuppressive treatment with high doses of intravenous methylprednisolone and cyclophosphamide and plasmapheresis, and required lung transplantation. Anti-MDA5 antibody titer declined and disappeared on anti-rejection treatment. These two cases underline the diagnostic conundrum and the therapeutic difficulties in patients with anti-MDA5 antibodies and clinically amyopathic dermatomyositis (CADM) or interstitial lung disease (ILD), who may undergo rapidly-progressive and fatal outcome. Presence of anti-MDA5 antibodies should always be suspected when confronted to CADM patients with cutaneous ulcerations or ILD to allow a rapid and adapted treatment initiation.

15.
Eur J Endocrinol ; 182(1): C1-C3, 2020 Jan.
Article En | MEDLINE | ID: mdl-31658441

In this issue of the European Journal of Endocrinology, Lin et al. report a de novo heterozygous nonsense variant in the PHEX gene in an X-linked hypophosphatemic rickets patient. The authors described a germline mosaicism in the sperm of the unaffected father of the proband, providing the opportunity to discuss the concept of isolated germline mosaicism. In addition to the genetic information passed on from generation to generation, each of us is born with a small number of novel genetic changes -de novo mutations- that occurred either prezygotically or postzygotically. When de novo mutational events occur prezygotically, mutation may pre-exist in a parent who is mosaic but the mutation might be inherited in the zygote and potentially in all cells of the developing offspring resulting in a de novo disease phenotype. Only somatic cells (mainly from blood and skin) are routinely used in genetic analyses, and a hint about possible germline mosaicism can be obtained only if somatic mosaicism is detected, or if more affected siblings are born with an identical de novo variant. There is now a wide range of disorders for which the occurrence of parental germline mosaicism has been reported. Interestingly, Lin et al. analyzed eight different tissues of the proband's father (including sperm), which allowed a conclusive assessment of his mosaicism extension. This study suggests that sperm analysis could more often be performed in our routine genetic screening for germline mosaicism in fathers of patients with apparently de novo mutations.


Mutation/genetics , Spermatozoa/metabolism , Fathers , Female , Genotype , Germ-Line Mutation/genetics , Heterozygote , Humans , Male , Mosaicism , Siblings
16.
Genes (Basel) ; 10(9)2019 08 22.
Article En | MEDLINE | ID: mdl-31443423

Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but high variable expressivity. NF1 is caused by loss-of-function mutations in the NF1 gene, a negative regulator of the RAS-MAPK pathway. The NF1 gene has one of the highest mutation rates in human disorders, which may explain the outbreak of independent de novo variants in the same family. Here, we report the co-occurrence of pathogenic variants in the NF1 and SPRED1 genes in six families with NF1 and Legius syndrome, using next-generation sequencing. In five of these families, we observed the co-occurrence of two independent NF1 variants. All NF1 variants were classified as pathogenic, according to the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG-AMP) guidelines. In the sixth family, one sibling inherited a complete deletion of the NF1 gene from her mother and carried a variant of unknown significance in the SPRED1 gene. This variant was also present in her brother, who was diagnosed with Legius syndrome, a differential diagnosis of NF1. This work illustrates the complexity of molecular diagnosis in a not-so-rare genetic disease.


Adaptor Proteins, Signal Transducing/genetics , Cafe-au-Lait Spots/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Phenotype , Adolescent , Adult , Cafe-au-Lait Spots/complications , Cafe-au-Lait Spots/pathology , Child , Female , Humans , Male , Mutation , Neurofibromatosis 1/complications , Neurofibromatosis 1/pathology , Pedigree
17.
Ann Biol Clin (Paris) ; 75(5): 589-594, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28958971

Two procedures for checking agreement between twin analyzers (Abbott Architect ci 8200) were tested in 23 blood and 7 urine parameters (10,160 paired results from 7,882 blood and 2,278 urine tests). Two protocols were compared. In protocol 1, acceptance criterion is based on standard-deviation originated either from French recommendations (Société française de biologie clinique) or from within subject biological variation. In protocol 2, acceptance criterion is based on values of expanded uncertainty of measurements calculated according to SH GTA 04. Percentages of comparisons refused were significantly different (p < 0.05) and varied from 0.0 to 18.6% (median: 0.45%) and from 0.0 to 8.75% (median: 1.2%). For 9 blood parameters (sodium, total CO2 , total proteins, calcium, urea, LDH, amylase, lipase and troponin I) and 4 urine parameters (sodium, protein, glucose, creatinine) a significant difference was observed between protocol 1 and 2. For the majority of tests, evaluation criterion based on within run standard deviation (protocol 1) is less stringent than protocol 2 based on expanded uncertainly of measurements. The use of expanded uncertainty as comparability criterion seems to be an interesting approach, especially for assays presenting wide theoretical physiopathology changes.


Blood Chemical Analysis/instrumentation , Clinical Laboratory Techniques/instrumentation , Urinalysis/instrumentation , Automation, Laboratory/instrumentation , Blood Chemical Analysis/methods , Creatinine/blood , Creatinine/urine , France , Humans , Reproducibility of Results , Sodium/blood , Sodium/urine , Urea/blood , Urea/urine , Urinalysis/methods
18.
Ann Biol Clin (Paris) ; 75(2): 222-224, 2017 Apr 01.
Article En | MEDLINE | ID: mdl-28377335

Type 1 diabetes accounts for 5 to 10% of all diabetes cases. It appears most often during childhood or adolescence. We report the case of a hypertensive overweight patient of 79 years who consulted for asthenia in context of polyuria-polydipsia syndrome in which blood glucose assay revealed a diabetes mellitus associated with ketosis. Despite the age, the body mass index and the history of the patient, type 1 diabetes diagnosis should be considered in this clinical presentation with brutal and symptomatic early. The imagery excluding a pancreatic tumor and positive research of circulating autoantibodies will confirm the diagnosis.


Diabetes Mellitus, Type 1/diagnosis , Hyperglycemia/diagnosis , Polydipsia/diagnosis , Polyuria/diagnosis , Age Factors , Aged , Diagnosis, Differential , Female , Humans , Hyperglycemia/complications , Late Onset Disorders , Polydipsia/complications , Polyuria/complications , Weight Loss/physiology
...