Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Genet ; 106(1): 47-55, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38378010

ABSTRACT

Skeletal dysplasias (SKDs) are a heterogeneous group of more than 750 genetic disorders characterized by abnormal development, growth, and maintenance of bones or cartilage in the human skeleton. SKDs are often caused by variants in early patterning genes and in many cases part of multiple malformation syndromes and occur in combination with non-skeletal phenotypes. The aim of this study was to investigate the underlying genetic cause of congenital SKDs in highly consanguineous Pakistani families, as well as in sporadic and familial SKD cases from India using multigene panel sequencing analysis. Therefore, we performed panel sequencing of 386 bone-related genes in 7 highly consanguineous families from Pakistan and 27 cases from India affected with SKDs. In the highly consanguineous families, we were able to identify the underlying genetic cause in five out of seven families, resulting in a diagnostic yield of 71%. Whereas, in the sporadic and familial SKD cases, we identified 12 causative variants, corresponding to a diagnostic yield of 44%. The genetic heterogeneity in our cohorts was very high and we were able to detect various types of variants, including missense, nonsense, and frameshift variants, across multiple genes known to cause different types of SKDs. In conclusion, panel sequencing proved to be a highly effective way to decipher the genetic basis of SKDs in highly consanguineous families as well as sporadic and or familial cases from South Asia. Furthermore, our findings expand the allelic spectrum of skeletal dysplasias.


Subject(s)
Consanguinity , Pedigree , Humans , Male , Female , Pakistan/epidemiology , India/epidemiology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/pathology , Phenotype , Child , Mutation , Bone Diseases, Developmental/genetics , Genetic Predisposition to Disease , Child, Preschool , High-Throughput Nucleotide Sequencing , Genetic Heterogeneity
2.
Clin Genet ; 105(6): 639-654, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38374498

ABSTRACT

The application of genomic technologies has led to unraveling of the complex genetic landscape of disorders of epilepsy, gaining insights into their underlying disease mechanisms, aiding precision medicine, and providing informed genetic counseling. We herein present the phenotypic and genotypic insights from 142 Indian families with epilepsy with or without comorbidities. Based on the electroclinical findings, epilepsy syndrome diagnosis could be made in 44% (63/142) of the families adopting the latest proposal for the classification by the ILAE task force (2022). Of these, 95% (60/63) of the families exhibited syndromes with developmental epileptic encephalopathy or progressive neurological deterioration. A definitive molecular diagnosis was achieved in 74 of 142 (52%) families. Infantile-onset epilepsy was noted in 81% of these families (61/74). Fifty-five monogenic, four chromosomal, and one imprinting disorder were identified in 74 families. The genetic variants included 65 (96%) single-nucleotide variants/small insertion-deletions, 1 (2%) copy-number variant, and 1 (2%) triplet-repeat expansion in 53 epilepsy-associated genes causing monogenic disorders. Of these, 35 (52%) variants were novel. Therapeutic implications were noted in 51% of families (38/74) with definitive diagnosis. Forty-one out of 66 families with monogenic disorders exhibited autosomal recessive and inherited autosomal dominant disorders with high risk of recurrence.


Subject(s)
Epilepsy , Genetic Counseling , Phenotype , Humans , Epilepsy/genetics , Epilepsy/epidemiology , Epilepsy/diagnosis , India/epidemiology , Male , Female , Child , Child, Preschool , Infant , Genetic Predisposition to Disease , Pedigree , Age of Onset , Genetic Association Studies , Adolescent , Genotype , DNA Copy Number Variations/genetics
3.
Am J Med Genet A ; 194(7): e63566, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38357848

ABSTRACT

PRKACA-related, atrial defects-polydactyly-multiple congenital malformation syndrome is a recently described skeletal ciliopathy, which is caused by disease-causing variants in PRKACA. The primary phenotypic description includes atrial septal defects, and limb anomalies including polydactyly and short limbs. To date, only four molecularly proven patients have been reported in the literature with a recurrent variant, c.409G>A p.Gly137Arg in PRKACA. In this study, we report the fifth affected individual with the same variant and review the clinical features and radiographic findings of this rare syndrome.


Subject(s)
Abnormalities, Multiple , Polydactyly , Humans , Polydactyly/genetics , Polydactyly/pathology , Polydactyly/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Abnormalities, Multiple/diagnosis , Female , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/diagnostic imaging , Heart Septal Defects, Atrial/diagnosis , Heart Septal Defects, Atrial/pathology , Male , Phenotype , Mutation/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/diagnostic imaging , India
4.
Nature ; 561(7722): E7, 2018 09.
Article in English | MEDLINE | ID: mdl-29977062

ABSTRACT

In this Letter, the surname of author Lena Vlaminck was misspelled 'Vlaeminck'. In addition, author Kris Vleminckx should have been associated with affiliation 16 (Center for Medical Genetics, Ghent University, Ghent, Belgium). These have been corrected online.

5.
Nature ; 557(7706): 564-569, 2018 05.
Article in English | MEDLINE | ID: mdl-29769720

ABSTRACT

The four R-spondin secreted ligands (RSPO1-RSPO4) act via their cognate LGR4, LGR5 and LGR6 receptors to amplify WNT signalling1-3. Here we report an allelic series of recessive RSPO2 mutations in humans that cause tetra-amelia syndrome, which is characterized by lung aplasia and a total absence of the four limbs. Functional studies revealed impaired binding to the LGR4/5/6 receptors and the RNF43 and ZNRF3 transmembrane ligases, and reduced WNT potentiation, which correlated with allele severity. Unexpectedly, however, the triple and ubiquitous knockout of Lgr4, Lgr5 and Lgr6 in mice did not recapitulate the known Rspo2 or Rspo3 loss-of-function phenotypes. Moreover, endogenous depletion or addition of exogenous RSPO2 or RSPO3 in triple-knockout Lgr4/5/6 cells could still affect WNT responsiveness. Instead, we found that the concurrent deletion of rnf43 and znrf3 in Xenopus embryos was sufficient to trigger the outgrowth of supernumerary limbs. Our results establish that RSPO2, without the LGR4/5/6 receptors, serves as a direct antagonistic ligand to RNF43 and ZNRF3, which together constitute a master switch that governs limb specification. These findings have direct implications for regenerative medicine and WNT-associated cancers.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Extremities/embryology , Intercellular Signaling Peptides and Proteins/metabolism , Limb Deformities, Congenital/genetics , Receptors, G-Protein-Coupled/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , DNA-Binding Proteins/metabolism , Female , Fibroblasts , Gene Knockout Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , Phenotype , Receptors, G-Protein-Coupled/deficiency , Ubiquitin-Protein Ligases/metabolism , Xenopus/genetics
6.
Am J Med Genet A ; 191(8): 2175-2180, 2023 08.
Article in English | MEDLINE | ID: mdl-37337996

ABSTRACT

Heterozygous disease-causing variants in BCL11B are the basis of a rare neurodevelopmental syndrome with craniofacial and immunological involvement. Isolated craniosynostosis, without systemic or immunological findings, has been reported in one of the 17 individuals reported with this disorder till date. We report three additional individuals harboring de novo heterozygous frameshift variants, all lying in the exon 4 of BCL11B. All three individuals presented with the common findings of this disorder i.e. developmental delay, recurrent infections with immunologic abnormalities and facial dysmorphism. Notably, craniosynostosis of variable degree was seen in all three individuals. We, thus add to the evolving genotypes and phenotypes of BCL11B-related BAFopathy and also review the clinical, genomic spectrum along with the underlying disease mechanisms of this disorder.


Subject(s)
Craniosynostoses , Intellectual Disability , Neurodevelopmental Disorders , Humans , Transcription Factors/genetics , Craniosynostoses/diagnosis , Craniosynostoses/genetics , Frameshift Mutation , Phenotype , Tumor Suppressor Proteins/genetics , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Repressor Proteins/genetics
7.
Brain ; 145(12): 4202-4209, 2022 12 19.
Article in English | MEDLINE | ID: mdl-35953447

ABSTRACT

Hypomyelinating leukodystrophies comprise a subclass of genetic disorders with deficient myelination of the CNS white matter. Here we report four unrelated families with a hypomyelinating leukodystrophy phenotype harbouring variants in TMEM163 (NM_030923.5). The initial clinical presentation resembled Pelizaeus-Merzbacher disease with congenital nystagmus, hypotonia, delayed global development and neuroimaging findings suggestive of significant and diffuse hypomyelination. Genomic testing identified three distinct heterozygous missense variants in TMEM163 with two unrelated individuals sharing the same de novo variant. TMEM163 is highly expressed in the CNS particularly in newly myelinating oligodendrocytes and was recently revealed to function as a zinc efflux transporter. All the variants identified lie in highly conserved residues in the cytoplasmic domain of the protein, and functional in vitro analysis of the mutant protein demonstrated significant impairment in the ability to efflux zinc out of the cell. Expression of the mutant proteins in an oligodendroglial cell line resulted in substantially reduced mRNA expression of key myelin genes, reduced branching and increased cell death. Our findings indicate that variants in TMEM163 cause a hypomyelinating leukodystrophy and uncover a novel role for zinc homeostasis in oligodendrocyte development and myelin formation.


Subject(s)
Pelizaeus-Merzbacher Disease , Humans , Pelizaeus-Merzbacher Disease/genetics , Mutation, Missense , Myelin Sheath/metabolism , Zinc/metabolism , Membrane Proteins/genetics
8.
Brain ; 145(6): 1916-1923, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35202461

ABSTRACT

The Kennedy pathways catalyse the de novo synthesis of phosphatidylcholine and phosphatidylethanolamine, the most abundant components of eukaryotic cell membranes. In recent years, these pathways have moved into clinical focus because four of ten genes involved have been associated with a range of autosomal recessive rare diseases such as a neurodevelopmental disorder with muscular dystrophy (CHKB), bone abnormalities and cone-rod dystrophy (PCYT1A) and spastic paraplegia (PCYT2, SELENOI). We identified six individuals from five families with bi-allelic variants in CHKA presenting with severe global developmental delay, epilepsy, movement disorders and microcephaly. Using structural molecular modelling and functional testing of the variants in a cell-based Saccharomyces cerevisiae model, we determined that these variants reduce the enzymatic activity of CHKA and confer a significant impairment of the first enzymatic step of the Kennedy pathway. In summary, we present CHKA as a novel autosomal recessive gene for a neurodevelopmental disorder with epilepsy and microcephaly.


Subject(s)
Choline Kinase , Epilepsy , Microcephaly , Nervous System Malformations , Neurodevelopmental Disorders , Alleles , Choline Kinase/genetics , Epilepsy/genetics , Humans , Microcephaly/complications , Microcephaly/genetics , Nervous System Malformations/genetics , Neurodevelopmental Disorders/genetics
9.
Am J Hum Genet ; 105(1): 132-150, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31230720

ABSTRACT

Arthrogryposis is a clinical finding that is present either as a feature of a neuromuscular condition or as part of a systemic disease in over 400 Mendelian conditions. The underlying molecular etiology remains largely unknown because of genetic and phenotypic heterogeneity. We applied exome sequencing (ES) in a cohort of 89 families with the clinical sign of arthrogryposis. Additional molecular techniques including array comparative genomic hybridization (aCGH) and Droplet Digital PCR (ddPCR) were performed on individuals who were found to have pathogenic copy number variants (CNVs) and mosaicism, respectively. A molecular diagnosis was established in 65.2% (58/89) of families. Eleven out of 58 families (19.0%) showed evidence for potential involvement of pathogenic variation at more than one locus, probably driven by absence of heterozygosity (AOH) burden due to identity-by-descent (IBD). RYR3, MYOM2, ERGIC1, SPTBN4, and ABCA7 represent genes, identified in two or more families, for which mutations are probably causative for arthrogryposis. We also provide evidence for the involvement of CNVs in the etiology of arthrogryposis and for the idea that both mono-allelic and bi-allelic variants in the same gene cause either similar or distinct syndromes. We were able to identify the molecular etiology in nine out of 20 families who underwent reanalysis. In summary, our data from family-based ES further delineate the molecular etiology of arthrogryposis, yielded several candidate disease-associated genes, and provide evidence for mutational burden in a biological pathway or network. Our study also highlights the importance of reanalysis of individuals with unsolved diagnoses in conjunction with sequencing extended family members.


Subject(s)
Arthrogryposis/genetics , Arthrogryposis/pathology , DNA Copy Number Variations , Genetic Markers , Genomics/methods , Multifactorial Inheritance/genetics , Mutation , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Connectin/genetics , Female , Gestational Age , Humans , Infant , Infant, Newborn , Male , Mosaicism , Pedigree , Ryanodine Receptor Calcium Release Channel/genetics , Vesicular Transport Proteins/genetics , Exome Sequencing , Young Adult
10.
Clin Genet ; 101(2): 221-232, 2022 02.
Article in English | MEDLINE | ID: mdl-34750818

ABSTRACT

Homozygous variants in PPP2R3C have been reported to cause a syndromic 46,XY complete gonadal dysgenesis phenotype with extragonadal manifestations (GDRM, MIM# 618419) in patients from four unrelated families, whereas heterozygous variants have been linked to reduced fertility with teratozoospermia (SPGF36, MIM# 618420) in male carriers. We present eight patients from four unrelated families of Turkish and Indian descent with three different germline homozygous PPP2R3C variants including a novel in-frame duplication (c.639_647dupTTTCTACTC, p.Ser216_Tyr218dup). All patients exhibit recognizable facial dysmorphisms allowing gestalt diagnosis. In two 46,XX patients with hypergonadotropic hypogonadism and nonvisualized gonads, primary amenorrhea along with absence of secondary sexual characteristics and/or unique facial gestalt led to the diagnosis. 46,XY affected individuals displayed a spectrum of external genital phenotypes from ambiguous genitalia to complete female. We expand the spectrum of syndromic PPP2R3C-related XY gonadal dysgenesis to both XY and XX gonadal dysgenesis. Our findings supported neither ocular nor muscular involvement as major criteria of the syndrome. We also did not encounter infertility problems in the carriers. Since both XX and XY individuals were affected, we hypothesize that PPP2R3C is essential in the early signaling cascades controlling sex determination in humans.


Subject(s)
Gonadal Dysgenesis, 46,XX/diagnosis , Gonadal Dysgenesis, 46,XX/genetics , Gonadal Dysgenesis, 46,XY/diagnosis , Gonadal Dysgenesis, 46,XY/genetics , Mutation , Phenotype , Protein Phosphatase 2/genetics , Abnormalities, Multiple/genetics , Consanguinity , Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Pedigree , Polymorphism, Single Nucleotide
11.
Hum Mol Genet ; 28(16): 2720-2737, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31042281

ABSTRACT

Mutations in genes encoding components of the intraflagellar transport (IFT) complexes have previously been associated with a spectrum of diseases collectively termed ciliopathies. Ciliopathies relate to defects in the formation or function of the cilium, a sensory or motile organelle present on the surface of most cell types. IFT52 is a key component of the IFT-B complex and ensures the interaction of the two subcomplexes, IFT-B1 and IFT-B2. Here, we report novel IFT52 biallelic mutations in cases with a short-rib thoracic dysplasia (SRTD) or a congenital anomaly of kidney and urinary tract (CAKUT). Combining in vitro and in vivo studies in zebrafish, we showed that SRTD-associated missense mutation impairs IFT-B complex assembly and IFT-B2 ciliary localization, resulting in decreased cilia length. In comparison, CAKUT-associated missense mutation has a mild pathogenicity, thus explaining the lack of skeletal defects in CAKUT case. In parallel, we demonstrated that the previously reported homozygous nonsense IFT52 mutation associated with Sensenbrenner syndrome [Girisha et al. (2016) A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy. Clin. Genet., 90, 536-539] leads to exon skipping and results in a partially functional protein. Finally, our work uncovered a novel role for IFT52 in microtubule network regulation. We showed that IFT52 interacts and partially co-localized with centrin at the distal end of centrioles where it is involved in its recruitment and/or maintenance. Alteration of this function likely contributes to centriole splitting observed in Ift52-/- cells. Altogether, our findings allow a better comprehensive genotype-phenotype correlation among IFT52-related cases and revealed a novel, extra-ciliary role for IFT52, i.e. disruption may contribute to pathophysiological mechanisms.


Subject(s)
Carrier Proteins/genetics , Centrosome/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Microtubules/metabolism , Mutation , Amino Acid Sequence , Animals , Animals, Genetically Modified , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Child , Child, Preschool , Cilia/metabolism , Consanguinity , DNA Mutational Analysis , Female , Genotype , Homozygote , Humans , Infant , Intracellular Signaling Peptides and Proteins , Male , Pedigree , Phenotype , Protein Binding , Protein Interaction Domains and Motifs/genetics , Trimethoprim, Sulfamethoxazole Drug Combination/metabolism , Exome Sequencing , Zebrafish
12.
Clin Genet ; 99(1): 119-132, 2021 01.
Article in English | MEDLINE | ID: mdl-33047326

ABSTRACT

Several genetic disorders have variable degree of central nervous system white matter abnormalities. We retrieved and reviewed 422 genetic conditions with prominent and consistent involvement of white matter from the literature. We herein describe the current definitions, classification systems, clinical spectrum, neuroimaging findings, genomics, and molecular mechanisms of these conditions. Though diagnosis for most of these disorders relies mainly on genomic tests, specifically exome sequencing, we collate several clinical and neuroimaging findings still relevant in diagnosis of clinically recognizable disorders. We also review the current understanding of pathophysiology and therapeutics of these disorders.


Subject(s)
Central Nervous System Diseases/genetics , Genomics , Leukoencephalopathies/genetics , Nervous System Malformations/genetics , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/diagnostic imaging , Humans , Leukoencephalopathies/diagnosis , Leukoencephalopathies/diagnostic imaging , Magnetic Resonance Imaging , Mass Screening , Nervous System Malformations/diagnosis , Nervous System Malformations/diagnostic imaging , White Matter/diagnostic imaging , White Matter/pathology , Exome Sequencing
13.
Clin Genet ; 99(4): 594-600, 2021 04.
Article in English | MEDLINE | ID: mdl-33463720

ABSTRACT

RNA exosome is a highly conserved ribonuclease complex essential for RNA processing and degradation. Bi-allelic variants in exosome subunits EXOSC3, EXOSC8 and EXOSC9 have been reported to cause pontocerebellar hypoplasia type 1B, type 1C and type 1D, respectively, while those in EXOSC2 cause short stature, hearing loss, retinitis pigmentosa and distinctive facies. We ascertained an 8-months-old male with developmental delay, microcephaly, subtle dysmorphism and hypotonia. Pontocerebellar hypoplasia and delayed myelination were noted on neuroimaging. A similarly affected elder sibling succumbed at the age of 4-years 6-months. Chromosomal microarray returned normal results. Exome sequencing revealed a homozygous missense variant, c.104C > T p.(Ser35Leu) in EXOSC1 (NM_016046.5) as the possible candidate. In silico mutagenesis revealed loss of a polar contact with neighboring Leu37 residue. Quantitative real-time PCR indicated no appreciable differences in EXOSC1 transcript levels. Immunoblotting and blue native PAGE revealed reduction in the EXOSC1 protein levels and EXO9 complex in the proband, respectively. We herein report an individual with the bi-allelic variant c.104C>T p.(Ser35Leu) in EXOSC1 and clinical features of pontocerebellar hypoplasia type 1. Immunoblotting and blue native PAGE provide evidence for the pathogenicity of the variant. Thus, we propose EXOSC1 as a novel candidate gene for pontocerebellar hypoplasia.


Subject(s)
Cerebellar Diseases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Mutation, Missense , RNA-Binding Proteins/genetics , Alleles , Amino Acid Sequence , Amino Acid Substitution , Brain/pathology , Consanguinity , Developmental Disabilities/genetics , Humans , Infant , Male , Pedigree , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Exome Sequencing
14.
Clin Genet ; 100(2): 201-205, 2021 08.
Article in English | MEDLINE | ID: mdl-33890291

ABSTRACT

The glycine cleavage system H protein (GCSH) is an integral part of the glycine cleavage system with its additional involvement in the synthesis and transport of lipoic acid. We hypothesize that pathogenic variants in GCSH can cause variant nonketotic hyperglycinemia (NKH), a heterogeneous group of disorders with findings resembling a combination of severe NKH (elevated levels of glycine in plasma and CSF, progressive lethargy, seizures, severe hypotonia, no developmental progress, early death) and mitochondriopathies (lactic acidosis, leukoencephalopathy and Leigh-like lesions on MRI). We herein report three individuals from two unrelated Indian families with clinical, biochemical, and radiological findings of variant NKH, harboring a biallelic start loss variant, c.1A > G in GCSH.


Subject(s)
Glycine Decarboxylase Complex H-Protein/genetics , Hyperglycinemia, Nonketotic/genetics , Child, Preschool , Female , Glycine/blood , Glycine/cerebrospinal fluid , Humans , Hyperglycinemia, Nonketotic/etiology , Male , Mutation , Pedigree
15.
Clin Genet ; 100(5): 542-550, 2021 11.
Article in English | MEDLINE | ID: mdl-34302356

ABSTRACT

Genetic disorders with predominant central nervous system white matter abnormalities (CNS WMAs), also called leukodystrophies, are heterogeneous entities. We ascertained 117 individuals with CNS WMAs from 104 unrelated families. Targeted genetic testing was carried out in 16 families and 13 of them received a diagnosis. Chromosomal microarray (CMA) was performed for three families and one received a diagnosis. Mendeliome sequencing was used for testing 11 families and all received a diagnosis. Whole exome sequencing (WES) was performed in 80 families and was diagnostic in 52 (65%). Singleton WES was diagnostic for 50/75 (66.67%) families. Overall, genetic diagnoses were obtained in 77 families (74.03%). Twenty-two of 47 distinct disorders observed in this cohort have not been reported in Indian individuals previously. Notably, disorders of nuclear mitochondrial pathology were most frequent (9 disorders in 20 families). Thirty-seven of 75 (49.33%) disease-causing variants are novel. To sum up, the present cohort describes the phenotypic and genotypic spectrum of genetic disorders with CNS WMAs in our population. It demonstrates WES, especially singleton WES, as an efficient tool in the diagnosis of these heterogeneous entities. It also highlights possible founder events and recurrent disease-causing variants in our population and their implications on the testing strategy.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , White Matter/abnormalities , Alleles , Chromosome Aberrations , Consanguinity , Family , Genetic Association Studies/methods , Genetic Testing , Humans , India/epidemiology , Microarray Analysis , Mutation , Nervous System Malformations/epidemiology , Exome Sequencing
16.
Am J Med Genet A ; 185(5): 1602-1605, 2021 05.
Article in English | MEDLINE | ID: mdl-33559318

ABSTRACT

Wiedemann-Rautenstrauch syndrome (WRS; MIM# 264090) is a rare neonatal progeroid disorder resulting from biallelic pathogenic variants in the POLR3A. It is an autosomal recessive condition characterized by growth retardation, lipoatrophy, a distinctive face, sparse scalp hair, and dental anomalies. Till date, 19 families are reported with WRS due to variants in POLR3A. Here, we describe an 18 months old male child with biallelic c.2005C>T p.(Arg669Ter) and c.1771-7C>G variant in heterozygous state identified by exome sequencing in POLR3A leading to WRS phenotype. The variant c.1771-7C>G was earlier found to be associated with hereditary spastic ataxia. We emphasize on the phenotype in an Indian patient with WRS.


Subject(s)
Fetal Growth Retardation/genetics , Genetic Predisposition to Disease , Progeria/genetics , RNA Polymerase III/genetics , Alleles , Female , Fetal Growth Retardation/mortality , Fetal Growth Retardation/pathology , Humans , Infant , Infant Mortality , Infant, Newborn , Male , Mutation/genetics , Phenotype , Progeria/mortality , Progeria/pathology , Exome Sequencing
17.
Ann Hum Genet ; 84(4): 345-351, 2020 07.
Article in English | MEDLINE | ID: mdl-32124427

ABSTRACT

Leigh syndrome is a clinically and radiologically heterogeneous condition with approximately 75 genes, nuclear and mitochondrial, known to be implicated in its pathogenesis. Leigh syndrome due to complex II deficiency constitutes 2% to 7% of these cases. Previously, nine individuals with Leigh syndrome have been reported with pathogenic variants in SDHB, which encodes for the iron-sulfur cluster subunit of mitochondrial respiratory chain complex II. The proband presented with Leigh syndrome. Exome sequencing revealed a homozygous missense variant p.(Ala102Thr) in SDHB. In silico protein modeling of the wild-type and mutant proteins showed potentially decreased protein stability. We hereby report another individual with Leigh syndrome due to SDHB-related mitochondrial complex II deficiency and review the phenotype and genotype associated with this condition.


Subject(s)
Electron Transport Complex II/deficiency , Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/genetics , Succinate Dehydrogenase/genetics , Electron Transport Complex II/genetics , Homozygote , Humans , Infant , Male , Mutation, Missense , Protein Structure, Secondary , Exome Sequencing
18.
Am J Med Genet A ; 182(10): 2226-2229, 2020 10.
Article in English | MEDLINE | ID: mdl-33043633

ABSTRACT

Trichothiodystrophy, non-photosensitive type 4 (TTD4), is a rare genetic disorder with an autosomal recessive mode of inheritance. It is characterized by coarse and brittle hair, anomalies of the tissues derived from the neuro-ectoderm (skin, hair, and nails) and intellectual disability. We herein report two male siblings aged 13 and 16 years with TTD4 and a known homozygous pathogenic variant, c.229del [p.(Arg77Glyfs*76)] in exon 1 of MPLKIP (NM_138701.3). We herein highlight the clinical and molecular findings of the first reported case of TTD4 in probands of Indian ethnicity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Hair Diseases/genetics , Intellectual Disability/genetics , Trichothiodystrophy Syndromes/genetics , Adolescent , Exons/genetics , Genetic Predisposition to Disease , Hair/pathology , Hair Diseases/pathology , Humans , Intellectual Disability/physiopathology , Male , Nails/pathology , Skin/pathology , Trichothiodystrophy Syndromes/pathology
19.
Am J Med Genet A ; 182(11): 2699-2703, 2020 11.
Article in English | MEDLINE | ID: mdl-32864817

ABSTRACT

Bi-allelic HOXA1 pathogenic variants clinically manifest as two distinct syndromes, Bosley-Salih-Alorainy syndrome (BSAS) and Athabascan brainstem dysgenesis syndrome, mainly reported in two different populations from Saudi Arabia and southwest North America, respectively. Here we report two siblings of Indian origin with BSAS phenotype caused by a novel homozygous exon 2 HOXA1 pathogenic variants.


Subject(s)
Brain Stem/abnormalities , Hearing Loss, Sensorineural/pathology , Homeodomain Proteins/genetics , Homozygote , Mutation , Nervous System Malformations/pathology , Ocular Motility Disorders/pathology , Phenotype , Transcription Factors/genetics , Adolescent , Adult , Brain Stem/pathology , Female , Hearing Loss, Sensorineural/genetics , Humans , India , Male , Nervous System Malformations/genetics , Ocular Motility Disorders/genetics , Young Adult
20.
Am J Hum Genet ; 98(6): 1243-1248, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27236923

ABSTRACT

Spondyloepimetaphyseal dysplasias (SEMDs) comprise a heterogeneous group of autosomal-dominant and autosomal-recessive disorders. An apparent X-linked recessive (XLR) form of SEMD in a single Italian family was previously reported. We have been able to restudy this family together with a second family from Korea by segregating a severe SEMD in an X-linked pattern. Exome sequencing showed missense mutations in BGN c.439A>G (p.Lys147Glu) in the Korean family and c.776G>T (p.Gly259Val) in the Italian family; the c.439A>G (p.Lys147Glu) mutation was also identified in a further simplex SEMD case from India. Biglycan is an extracellular matrix proteoglycan that can bind transforming growth factor beta (TGF-ß) and thus regulate its free concentration. In 3-dimensional simulation, both altered residues localized to the concave arc of leucine-rich repeat domains of biglycan that interact with TGF-ß. The observation of recurrent BGN mutations in XLR SEMD individuals from different ethnic backgrounds allows us to define "XLR SEMD, BGN type" as a nosologic entity.


Subject(s)
Biglycan/genetics , Genetic Diseases, X-Linked/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , Adult , Aged , Amino Acid Sequence , Biglycan/chemistry , Biglycan/metabolism , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Pedigree , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL