Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1345067, 2024.
Article in English | MEDLINE | ID: mdl-38544690

ABSTRACT

Background: Mandibuloacral dysplasia (MAD) syndrome is a rare genetic disease. Several progeroid syndromes including mandibuloacral dysplasia type A (MADA), mandibuloacral dysplasia type B(MADB), Hutchinson-Gilford progeria (HGPS) and mandibular hypoplasia, deafness, and lipodystrophy syndrome (MDPL) have been reported previously. A novel MAD progeroid syndrome (MADaM) has recently been reported. So far, 7 cases of MADaM diagnosed with molecular diagnostics have been reported in worldwide. In the Chinese population, cases of MAD associated with the MTX2 variant have never been reported. Methods: The clinical symptoms and the genetic analysis were identified and investigated in patients presented with the disease. In addition, we analyzed and compared 7 MADaM cases reported worldwide and summarized the progeroid syndromes reported in the Chinese population to date. Results: The present study reports a case of a novel homozygous mutation c.378 + 1G > A in the MTX2 gene, which has not been previously reported in the literature. Patients present with early onset and severe symptoms and soon after birth are found to have growth retardation. In addition to the progeroid features, skeletal deformities, generalized lipodystrophy reported previously, and other multisystem involvement, e.g. hepatosplenic, renal, and cardiovascular system, this case was also reported to have combined hypogammaglobulinemia. She has since been admitted to the hospital several times for infections. Among 22 previously reported progeroid syndromes, 16/22 were MADA or HGPS caused by LMNA gene mutations, and the homozygous c.1579C > T (p.R527C) mutation may be a hot spot mutation for MAD in the Chinese population. MAD and HGPS mostly present in infancy with skin abnormalities or alopecia, MDPL mostly presents in school age with growth retardation as the first manifestation, and is often combined with an endocrine metabolism disorder after several decades. Conclusion: This is the first case of MAD syndrome caused by mutations in MTX2 gene reported in the Chinese population. MTX2 gene c.378 + 1G > A homozygous mutation has not been previously reported and the report of this patient expands the spectrum of MTX2 mutations. In addition, we summarized the genotypes and clinical characteristics of patients with progeroid syndromes in China.


Subject(s)
Lipodystrophy , Progeria , Female , Humans , Progeria/genetics , Progeria/complications , Progeria/diagnosis , Lipodystrophy/genetics , Syndrome , Mutation , Rare Diseases , Growth Disorders/complications
2.
Genes (Basel) ; 15(2)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38397171

ABSTRACT

Bi-allelic pathogenic variations within POLR3A have been associated with a spectrum of hereditary disorders. Among these, a less frequently observed condition is Wiedemann-Rautenstrauch syndrome (WRS), also known as neonatal progeroid syndrome. This syndrome typically manifests neonatally and is characterized by growth retardation, evident generalized lipodystrophy with distinctively localized fat accumulations, sparse scalp hair, and atypical facial features. Our objective was to elucidate the underlying molecular mechanisms of Wiedemann-Rautenstrauch syndrome (WRS). In this study, we present a clinical case of a 7-year-old female patient diagnosed with WRS. Utilizing whole-exome sequencing (WES), we identified a novel missense variant c.3677T>C (p.Leu1226Pro) in the POLR3A gene (NM_007055.4) alongside two cis intronic variants c.1909+22G>A and c.3337-11T>C. Via the analysis of mRNA derived from fibroblasts, we reconfirmed the splicing-affecting nature of the c.3337-11T>C variant. Furthermore, our investigation led to the reclassification of the c.3677T>C (p.Leu1226Pro) variant as a likely pathogenic variant. Therefore, this is the first case demonstrating the molecular genetics of a patient with Wiedemann-Rautenstrauch syndrome from the Russian Federation. A limited number of clinical cases have been documented until this moment; therefore, broadening the linkage between phenotype and molecular changes in the POLR3A gene will significantly contribute to the comprehensive understanding of the molecular basis of POLR3A-related disorders.


Subject(s)
Progeria , Infant, Newborn , Female , Humans , Child , Progeria/genetics , Progeria/diagnosis , Progeria/pathology , Fetal Growth Retardation/pathology , Mutation , Russia , RNA Polymerase III/genetics
5.
Circulation ; 147(23): 1734-1744, 2023 06 06.
Article in English | MEDLINE | ID: mdl-36919608

ABSTRACT

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare, fatal, premature aging disease caused by a toxic protein called progerin. Circulating progerin has not been previously detected, precluding research using readily available biological samples. This study aimed to develop a plasma progerin assay to evaluate progerin's quantity, response to progerin-targeted therapy, and relationship to patient survival. METHODS: Biological samples were collected by The Progeria Research Foundation Cell and Tissue Bank from a non-HGPS cohort cross-sectionally and a HGPS cohort longitudinally. HGPS donations occurred at baseline and intermittently while treated with farnesylation inhibitors lonafarnib±pravastatin and zoledronate, within 3 sequential open-label clinical trials at Boston Children's Hospital totaling >10 years of treatment. An ultrasensitive single-molecule counting progerin immunoassay was developed with prespecified performance parameters. Intra- and interpatient group statistics were descriptive. The relationship between progerin and survival was assessed by using joint modeling with time-dependent slopes parameterization. RESULTS: The assay's dynamic detection range was 59 to 30 000 pg/mL (R2=0.9987). There was no lamin A cross-reactivity. Mean plasma progerin in non-HGPS participants (n=69; 39 male, 30 female; age, 0.2-71.3 years) was 351±251 pg/mL, and in drug-naive participants with HGPS (n=74; 37 female, 37 male; age, 2.1-17.5 years) was 33 261±12 346 pg/mL, reflecting a 95-fold increase in affected children (P<0.0001). Progerin levels did not differ by sex (P=0.99). Lonafarnib treatment resulted in an average per-visit progerin decrease from baseline of between 35% to 62% (all P<0.005); effects were not augmented by adding pravastatin and zoledronate. Progerin levels fell within 4 months of therapy and remained lower for up to 10 years. The magnitude of progerin decrease positively associated with patient survival (P<0.0001; ie, 15 000 pg/mL decrease yields a 63.9% decreased risk of death). For any given decrease in progerin, life expectancy incrementally increased with longer treatment duration. CONCLUSIONS: A sensitive, quantitative immunoassay for progerin was developed and used to demonstrate high progerin levels in HGPS plasma that decreased with lonafarnib therapy. The extent of improved survival was associated with both the magnitude of progerin decrease and duration at lower levels. Thus, plasma progerin is a biomarker for HGPS whose reduction enables short- and long-term assessment of progerin-targeted treatment efficacy. REGISTRATION: URL: https://www. CLINICALTRIALS: gov. Unique identifiers: NCT00879034 and NCT00916747.


Subject(s)
Progeria , Child , Humans , Male , Female , Infant , Child, Preschool , Adolescent , Young Adult , Adult , Middle Aged , Aged , Progeria/diagnosis , Progeria/drug therapy , Progeria/metabolism , Zoledronic Acid/therapeutic use , Pravastatin/therapeutic use , Piperidines/therapeutic use , Lamin Type A/metabolism
6.
Probl Endokrinol (Mosk) ; 70(2): 86-93, 2023 Oct 08.
Article in Russian | MEDLINE | ID: mdl-38796765

ABSTRACT

Wiedemann-Rautenstrauch syndrome (neonatal progeroid syndrome) is an ultra-orphan disease from the group of premature aging syndromes with an autosomal recessive type of inheritance associated with mutations in the POLR3A, POLR3B, and POLR3GL genes encoding RNA polymerase III. The incidence of the disease is currently unknown. We present the first clinical description in Russian Federation of a patient 7 years 6 months old with Wiedemann-Rautenstrauch syndrome (compound heterozygous mutations in POLR3A gene) with progeroid features, adentia, growth retardation (height SDS -3,41, height velocity SDS -2,47), underweight (BMI SDS -6,20), and generalized lipodystrophy. The article presents the observation of the patient for 1.5 years, the world experience of dynamic follow-up of patients with neonatal progeroid syndrome, differential diagnosis, as well as recommendations for the management of patients with this syndrome. Given the lack of specific treatment to date, patients are observed by a multidisciplinary team of physicians.


Subject(s)
Progeria , Humans , Progeria/genetics , Progeria/diagnosis , Progeria/pathology , Child , RNA Polymerase III/genetics , Male , Female , Russia/epidemiology , Diagnosis, Differential , Mutation , Fetal Growth Retardation
8.
BMC Pediatr ; 22(1): 135, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35287606

ABSTRACT

BACKGROUND: Strokes in the paediatric age group have their own epidemiology and aetiology and are frequently misdiagnosed. As in the adult population, they present some risk factors that must be identified. Cerebral arteriopathies as a cause of paediatric ischaemic stroke present a very diverse aetiology and morphology. In this article we report a paediatric stroke in a patient who was diagnosed during his first months of life of Hutchinson-Gilford´s Progeria (HGP). This is a rare genetic condition caused by mutations in the LMNA gene, producing an aberrant lamin A protein. The disease leads to premature aging, and cardiovascular complications are the first cause of morbidity and mortality in these patients. CASE PRESENTATION: We report the case of a 5-year-old patient with HGP (missense mutation-de novo-c.1822G > A in heterozygosis, LMNA gene). The patient was diagnosed during his first year of life and presented distinct phenotypical features. No other relevant comorbidities were present. He was admitted to the emergency department for right hemiparesis with at least 4 h of evolution, with inability to open the hand and slight decrease in the level of consciousness (pedNIHSS 5-6). Cranial-CT and angio-CT showed findings indicative of left carotid dissection. Consensus was reached on conservative medical management with anticoagulation and antiplatelet therapy. In the first few days, the patient had a favourable evolution with resolution of the right lower limb hemiparesis and, one month after discharge, of the hand paresis. CONCLUSIONS: The clinical manifestations, the vascular phenotype of the genetic mutation and the location of the radiological signs on a specific vascular morphology are indicative of carotid dissection. Spontaneous dissections occur under a predisposing risk factor or disease and are an exceptional finding in patients with HGP.


Subject(s)
Brain Ischemia , Progeria , Stroke , Carotid Arteries , Child , Humans , Lamin Type A/genetics , Male , Mutation , Paresis , Progeria/diagnosis , Progeria/genetics , Rare Diseases , Stroke/etiology
10.
Clin Genet ; 99(1): 3-28, 2021 01.
Article in English | MEDLINE | ID: mdl-32860237

ABSTRACT

Progeroid disorders make up a heterogeneous group of very rare hereditary diseases characterized by clinical signs that often mimic physiological aging in a premature manner. Apart from Hutchinson-Gilford progeria syndrome, one of the best-investigated progeroid disorders, a wide spectrum of other premature aging phenotypes exist, which differ significantly in their clinical presentation and molecular pathogenesis. Next-generation sequencing (NGS)-based approaches have made it feasible to determine the molecular diagnosis in the early stages of a disease. Nevertheless, a broad clinical knowledge on these disorders and their associated symptoms is still fundamental for a comprehensive patient management and for the interpretation of variants of unknown significance from NGS data sets. This review provides a detailed overview on characteristic clinical features and underlying molecular genetics of well-known as well as only recently identified premature aging disorders and also highlights novel findings towards future therapeutic options.


Subject(s)
Aging, Premature/genetics , Aging/genetics , Progeria/genetics , Aging/pathology , Aging, Premature/diagnosis , Aging, Premature/physiopathology , High-Throughput Nucleotide Sequencing , Humans , Pathology, Molecular , Phenotype , Progeria/diagnosis , Progeria/physiopathology
11.
Am J Med Genet A ; 182(10): 2399-2402, 2020 10.
Article in English | MEDLINE | ID: mdl-32783369

ABSTRACT

Néstor-Guillermo progeria syndrome (NGPS; OMIM 614008) is characterized by early onset and slow progression of symptoms including poor growth, lipoatrophy, pseudosenile facial appearance, and normal cognitive development. In contrast to other progeria syndromes, NGPS is associated with a longer lifespan and higher risk for developing severe skeletal abnormalities. It is an autosomal recessive condition caused by biallelic pathogenic variants in BANF1. There are two previously reported patients with NGPS, both Spanish with molecular diagnoses made in adulthood and having the same homozygous pathogenic variant c.34G > A; p.Ala12Thr. Presented here is a 2 year, 8 month old girl with short stature, poor weight gain, sparse hair, and dysmorphic facial features reminiscent of premature aging. Whole exome sequencing identified the same c.34G > A homozygous pathogenic variant in BANF1 as reported in the previous patients. This is the first reported case of a child and is supporting evidence for this recurrent loss of function variant.


Subject(s)
Aging, Premature/genetics , DNA-Binding Proteins/genetics , Progeria/genetics , Adult , Aging, Premature/diagnosis , Aging, Premature/diagnostic imaging , Aging, Premature/pathology , Child , Child, Preschool , Female , Humans , Infant , Mutation/genetics , Phenotype , Progeria/diagnosis , Progeria/diagnostic imaging , Progeria/pathology , Exome Sequencing
12.
Nepal J Ophthalmol ; 12(23): 113-138, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32799251

ABSTRACT

INTRODUCTION: Progeria also known as Hutchinson Gilford Progeria Syndrome (HGPS) (MIM176670) is a very uncommon fatal genetic untimely aging syndrome. It is characterized by retarded physical development, accelerated degeneration of the skin, cardiovascular and musculoskeletal abnormalities. Other features include prominent eyes, thin nose, small chin and thin lips. Eyebrow hair loss, madarosis and lagopththalmos are the common ocular manifestations. CASE: We report a case of five year old boy with complaints of discomfort in bright light and a whitish appearance in his right eye for two months. He was accompanied by the parents. They complained of loss of eyelashes and eyebrows. In the developmental history he was normal at birth till the age of one year then they noticed gradual hair fall, delayed growth, wrinkling of skin, increase in size of head and thinning of limbs. CONCLUSION: This is the first case report from Nepal with the ocular presentation of progeria indicating the role of ocular senescence in patients with Hutchinson Progeria Gilford Syndrome.


Subject(s)
Progeria , Alopecia , Child, Preschool , Eye , Humans , Male , Nepal , Progeria/diagnosis , Progeria/genetics , Skin
14.
Calcif Tissue Int ; 106(3): 325-330, 2020 03.
Article in English | MEDLINE | ID: mdl-31807803

ABSTRACT

Mutations in the gene LMNA cause a wide spectrum of diseases that are now referred to laminopathies, such as muscular dystrophies, cardiomyopathies, and progeroid syndromes. Atypical progeroid syndrome (APS) is a type of progeroid syndrome mainly associated with LMNA mutations. Abnormal skeletal features associated with APS, such as osteoporosis and acroosteolysis, are rarely reported, and recurrent fractures have never been documented. We present a 16-year-old Chinese male patient with the typical features of APS, such as progeroid manifestations, cutaneous mottled hyperpigmentation, generalized lipodystrophy, and severe metabolic complications. The patient has also been detected with some rare and severe skeletal features, such as severe osteoporosis, generalized thinning of cortical bone, and recurrent femoral fractures. Genetic mutation detection in the LMNA gene revealed a de novo heterozygous mutation, the c. 29C>T (p. T10I).


Subject(s)
Femoral Fractures/diagnosis , Progeria/complications , Adolescent , Femoral Fractures/complications , Femoral Fractures/diagnostic imaging , Humans , Male , Progeria/diagnosis , Recurrence , Syndrome
15.
J Clin Res Pediatr Endocrinol ; 12(3): 315-318, 2020 09 02.
Article in English | MEDLINE | ID: mdl-31752481

ABSTRACT

Progeria syndrome is a rare disorder in childhood which causes accelerated systemic aging. Due to the accelerated aging process, disorders which normally occur only in old age will appear in these children at a much younger age. We report two children with progeria syndrome, in whom fulminant diabetes mellitus manifested at a very early age.


Subject(s)
Diabetes Mellitus/etiology , Progeria/complications , Progeria/diagnosis , Adolescent , Age Factors , Age of Onset , Child , DNA Repair-Deficiency Disorders/complications , DNA Repair-Deficiency Disorders/diagnosis , Diabetes Mellitus/diagnosis , Fatal Outcome , Female , Humans , Male
16.
J Dermatol Sci ; 96(2): 58-65, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31727429

ABSTRACT

Aging is an inevitable consequence of human life resulting in a gradual deterioration of cell, tissue and organismal function and an increased risk to develop chronic ailments. Premature aging syndromes, also known as progeroid syndromes, recapitulate many clinical features of normal aging and offer a unique opportunity to elucidate fundamental mechanisms that contribute to human aging. Progeroid syndromes can be broadly classified into those caused by perturbations of the nuclear lamina, a meshwork of proteins located underneath the inner nuclear membrane (laminopathies); and a second group that is caused by mutations that directly impair DNA replication and repair. We will focus mainly on laminopathies caused by incorrect processing of lamin A, an intermediate filament protein that resides at the nuclear periphery. Hutchinson-Gilford Progeria (HGPS) is an accelerated aging syndrome caused by a mutation in lamin A and one of the best studied laminopathies. HGPS patients exhibit clinical characteristics of premature aging, including alopecia, aberrant pigmentation, loss of subcutaneous fat and die in their teens as a result of atherosclerosis and cardiovascular complications. Here we summarize how cell- and mouse-based disease models provided mechanistic insights into human aging and discuss experimental strategies under consideration for the treatment of these rare genetic disorders.


Subject(s)
Aging, Premature/diagnosis , Aging, Premature/genetics , Lamin Type A/genetics , Nuclear Lamina/metabolism , Aging , Animals , Cell Nucleus/metabolism , Cellular Senescence , Chromatin/metabolism , Contracture/congenital , Contracture/diagnosis , Contracture/genetics , DNA Damage , DNA Repair , DNA Replication , Heterochromatin , Humans , Mice , Mutation , Nuclear Proteins/metabolism , Progeria/diagnosis , Progeria/genetics , Protein Precursors/genetics , Skin Abnormalities/diagnosis , Skin Abnormalities/genetics , Telomere/metabolism , Werner Syndrome/diagnosis , Werner Syndrome/genetics
18.
J Hum Genet ; 64(7): 609-616, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31015584

ABSTRACT

Individuals affected with autosomal recessive cutis laxa type 2B and 3 usually show translucent skin with visible veins and abnormal elastic fibers, intrauterine and/or postnatal growth restriction and a typical triangular facial gestalt. Here we describe three unrelated individuals in whom such a cutis laxa syndrome was suspected, especially after electron microscopy revealed immature and less dense dermal elastic fibers in one of them. However, one of these children also displayed optic atrophy and two hypogammaglobulinemia. All had elevated liver enzymes and acute liver failure during febrile episodes leading to early demise in two of them. The only surviving patient had been treated with immunoglobulins. Through exome sequencing we identified mutations in NBAS, coding for a protein involved in Golgi-to-ER transport. NBAS deficiency causes several rare conditions ranging from isolated recurrent acute liver failure to a multisystem disorder mainly characterized by short stature, optic nerve atrophy and Pelger-Huët anomaly (SOPH). Since we subsequently verified Pelger-Huët anomaly in two of the patients the diagnosis SOPH syndrome was unequivocally proven. Our data show that SOPH syndrome can be regarded as a differential diagnosis for the progeroid forms of cutis laxa in early infancy and that possibly treatment of the hypogammaglobulinemia can be of high relevance for the prognosis.


Subject(s)
Growth Disorders/diagnosis , Neoplasm Proteins/genetics , Optic Nerve Diseases/diagnosis , Pelger-Huet Anomaly/diagnosis , Agammaglobulinemia/blood , Agammaglobulinemia/physiopathology , Cutis Laxa/diagnosis , Cutis Laxa/genetics , Cutis Laxa/pathology , Diagnosis, Differential , Elastic Tissue/ultrastructure , Growth Disorders/genetics , Growth Disorders/pathology , Humans , Infant , Liver/enzymology , Liver/pathology , Male , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Pelger-Huet Anomaly/genetics , Pelger-Huet Anomaly/pathology , Progeria/diagnosis , Progeria/genetics , Skin/pathology , Syndrome , Exome Sequencing , Young Adult
19.
Hum Genet ; 137(11-12): 921-939, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30450527

ABSTRACT

Juvenile segmental progeroid syndromes are rare, heterogeneous disorders characterized by signs of premature aging affecting more than one tissue or organ starting in childhood. Hutchinson-Gilford progeria syndrome (HGPS), caused by a recurrent de novo synonymous LMNA mutation resulting in aberrant splicing and generation of a mutant product called progerin, is a prototypical example of such disorders. Here, we performed a joint collaborative study using massively parallel sequencing and targeted Sanger sequencing, aimed at delineating the underlying genetic cause of 14 previously undiagnosed, clinically heterogeneous, non-LMNA-associated juvenile progeroid patients. The molecular diagnosis was achieved in 11 of 14 cases (~ 79%). Furthermore, we firmly establish biallelic mutations in POLR3A as the genetic cause of a recognizable, neonatal, Wiedemann-Rautenstrauch-like progeroid syndrome. Thus, we suggest that POLR3A mutations are causal for a portion of under-diagnosed early-onset segmental progeroid syndromes. We additionally expand the clinical spectrum associated with PYCR1 mutations by showing that they can somewhat resemble HGPS in the first year of life. Moreover, our results lead to clinical reclassification in one single case. Our data emphasize the complex genetic and clinical heterogeneity underlying progeroid disorders.


Subject(s)
Fetal Growth Retardation/genetics , Progeria/genetics , Pyrroline Carboxylate Reductases/genetics , RNA Polymerase III/genetics , Adolescent , Alternative Splicing/genetics , Child , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/pathology , Genetic Predisposition to Disease , Humans , Infant , Lamin Type A/genetics , Male , Mutation , Phenotype , Progeria/diagnosis , Progeria/pathology , Progeria/physiopathology , delta-1-Pyrroline-5-Carboxylate Reductase
20.
J Med Genet ; 55(12): 837-846, 2018 12.
Article in English | MEDLINE | ID: mdl-30323018

ABSTRACT

BACKGROUND: Wiedemann-Rautenstrauch syndrome (WRS) is a form of segmental progeria presenting neonatally, characterised by growth retardation, sparse scalp hair, generalised lipodystrophy with characteristic local fatty tissue accumulations and unusual face. We aimed to understand its molecular cause. METHODS: We performed exome sequencing in two families, targeted sequencing in 10 other families and performed in silico modelling studies and transcript processing analyses to explore the structural and functional consequences of the identified variants. RESULTS: Biallelic POLR3A variants were identified in eight affected individuals and monoallelic variants of the same gene in four other individuals. In the latter, lack of genetic material precluded further analyses. Multiple variants were found to affect POLR3A transcript processing and were mostly located in deep intronic regions, making clinical suspicion fundamental to detection. While biallelic POLR3A variants have been previously reported in 4H syndrome and adolescent-onset progressive spastic ataxia, recurrent haplotypes specifically occurring in individuals with WRS were detected. All WRS-associated POLR3A amino acid changes were predicted to perturb substantially POLR3A structure/function. CONCLUSION: Biallelic mutations in POLR3A, which encodes for the largest subunit of the DNA-dependent RNA polymerase III, underlie WRS. No isolated functional sites in POLR3A explain the phenotype variability in POLR3A-related disorders. We suggest that specific combinations of compound heterozygous variants must be present to cause the WRS phenotype. Our findings expand the molecular mechanisms contributing to progeroid disorders.


Subject(s)
Alleles , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation/genetics , Progeria/diagnosis , Progeria/genetics , RNA Polymerase III/genetics , Adult , Amino Acid Sequence , Base Sequence , Computational Biology , Consanguinity , Female , Genotype , Haplotypes , Humans , Male , Models, Molecular , Mutation , Pedigree , Protein Conformation , RNA Polymerase III/chemistry , Reproducibility of Results , Sequence Analysis, DNA , Structure-Activity Relationship , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...