1,402 results on '"disease gene identification"'
Search Results
2. Scalable Non-Linear Graph Fusion for Prioritizing Cancer-Causing Genes.
- Author
-
Shah, Ekta and Maji, Pradipta
- Abstract
In the past few decades, both gene expression data and protein-protein interaction (PPI)networks have been extensively studied, due to their ability to depict important characteristics of disease-associated genes. In this regard, the paper presents a new gene prioritization algorithm to identify and prioritize cancer-causing genes, integrating judiciously the complementary information obtained from two data sources. The proposed algorithm selects disease-causing genes by maximizing the importance of selected genes and functional similarity among them. A new quantitative index is introduced to evaluate the importance of a gene. It considers whether a gene exhibits a differential expression pattern across sick and healthy individuals, and has a strong connectivity in the PPI network, which are the important characteristics of a potential biomarker. As disease-associated genes are expected to have similar expression profiles and topological structures, a scalable non-linear graph fusion technique, termed as ScaNGraF, is proposed to learn a disease-dependent functional similarity network from the co-expression and common neighbor based similarity networks. The proposed ScaNGraF, which is based on message passing algorithm, efficiently combines the shared and complementary information provided by different data sources with significantly lower computational cost. A new measure, termed as DiCoIN, is introduced to evaluate the quality of a learned affinity network. The performance of the proposed graph fusion technique and gene selection algorithm is extensively compared with that of some existing methods, using several cancer data sets. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
3. Decision Tree Based Model for the Classification of Pathogenic Gene Sequences Causing ASD
- Author
-
Sudha, V. Pream, Vijaya, M. S., Barbosa, Simone Diniz Junqueira, Series Editor, Filipe, Joaquim, Series Editor, Kotenko, Igor, Series Editor, Sivalingam, Krishna M., Series Editor, Washio, Takashi, Series Editor, Yuan, Junsong, Series Editor, Zhou, Lizhu, Series Editor, Deshpande, A.V., editor, Unal, Aynur, editor, Passi, Kalpdrum, editor, Singh, Dharm, editor, Nayak, Malaya, editor, Patel, Bharat, editor, and Pathan, Shafi, editor
- Published
- 2018
- Full Text
- View/download PDF
4. Exome sequencing in 38 patients with intracranial aneurysms and subarachnoid hemorrhage.
- Author
-
Sauvigny, Thomas, Alawi, Malik, Krause, Linda, Renner, Sina, Spohn, Michael, Busch, Alice, Kolbe, Verena, Altmüller, Janine, Löscher, Britt-Sabina, Franke, Andre, Brockmann, Christian, Lieb, Wolfgang, Westphal, Manfred, Schmidt, Nils Ole, Regelsberger, Jan, and Rosenberger, Georg
- Subjects
- *
INTRACRANIAL aneurysms , *SUBARACHNOID hemorrhage , *POPULATION genetics , *ETIOLOGY of diseases , *GENE frequency - Abstract
Objective: Genetic risk factors for unruptured intracranial aneurysms (UIA) and aneurysmal subarachnoid hemorrhage (aSAH) are poorly understood. We aimed to verify recently reported risk genes and to identify novel sequence variants involved in the etiology of UIA/aSAH. Methods: We performed exome sequencing (ES) in 35 unrelated individuals and 3 family members, each with a history of UIA and/or aSAH. We searched for sequence variants with minor allele frequency (MAF) ≤ 5% in the reported risk genes ADAMTS15, ANGPTL6, ARHGEF17, LOXL2, PCNT, RNF213, THSD1 and TMEM132B. To identify novel putative risk genes we looked for unknown (MAF = 0) variants shared by the three relatives. Results: We identified 20 variants with MAF ≤ 5% in 18 individuals: 9 variants in PCNT (9 patients), 4 in RNF213 (3 patients), 3 in THSD1 (6 patients), 2 in ANGPTL6 (3 patients), 1 in ADAMTS15 (1 patient) and 1 in TMEM132B (1 patient). In the affected family, prioritization of shared sequence variants yielded five novel putative risk genes. Based on predicted pathogenicity of identified variants, population genetics data and a high functional relevance for vascular biology, EDIL3 was selected as top candidate and screened in additional 37 individuals with UIA and/or aSAH: a further very rare EDIL3 sequence variant in two unrelated sporadic patients was identified. Conclusions: Our data support a role of sequence variants in PCNT, RNF213 and THSD1 as susceptibility factors for cerebrovascular disease. The documented function in vascular wall integrity, the crucial localization of affected amino acids and gene/variant association tests suggest EDIL3 as a further valid candidate disease gene for UIA/aSAH. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. Constructing an integrated gene similarity network for the identification of disease genes
- Author
-
Zhen Tian, Maozu Guo, Chunyu Wang, LinLin Xing, Lei Wang, and Yin Zhang
- Subjects
Gene Ontology ,Gene similarity networks ,Similarity network fusion ,Disease gene identification ,Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
Abstract Background Discovering novel genes that are involved human diseases is a challenging task in biomedical research. In recent years, several computational approaches have been proposed to prioritize candidate disease genes. Most of these methods are mainly based on protein-protein interaction (PPI) networks. However, since these PPI networks contain false positives and only cover less half of known human genes, their reliability and coverage are very low. Therefore, it is highly necessary to fuse multiple genomic data to construct a credible gene similarity network and then infer disease genes on the whole genomic scale. Results We proposed a novel method, named RWRB, to infer causal genes of interested diseases. First, we construct five individual gene (protein) similarity networks based on multiple genomic data of human genes. Then, an integrated gene similarity network (IGSN) is reconstructed based on similarity network fusion (SNF) method. Finally, we employee the random walk with restart algorithm on the phenotype-gene bilayer network, which combines phenotype similarity network, IGSN as well as phenotype-gene association network, to prioritize candidate disease genes. We investigate the effectiveness of RWRB through leave-one-out cross-validation methods in inferring phenotype-gene relationships. Results show that RWRB is more accurate than state-of-the-art methods on most evaluation metrics. Further analysis shows that the success of RWRB is benefited from IGSN which has a wider coverage and higher reliability comparing with current PPI networks. Moreover, we conduct a comprehensive case study for Alzheimer’s disease and predict some novel disease genes that supported by literature. Conclusions RWRB is an effective and reliable algorithm in prioritizing candidate disease genes on the genomic scale. Software and supplementary information are available at http://nclab.hit.edu.cn/~tianzhen/RWRB/ .
- Published
- 2017
- Full Text
- View/download PDF
6. Identifying Disease-Gene Associations With Graph-Regularized Manifold Learning
- Author
-
Ping Luo, Qianghua Xiao, Pi-Jing Wei, Bo Liao, and Fang-Xiang Wu
- Subjects
disease gene identification ,manifold learning ,disease module theory ,gene ontology ,multi-task learning ,Genetics ,QH426-470 - Abstract
Complex diseases are known to be associated with disease genes. Uncovering disease-gene associations is critical for diagnosis, treatment, and prevention of diseases. Computational algorithms which effectively predict candidate disease-gene associations prior to experimental proof can greatly reduce the associated cost and time. Most existing methods are disease-specific which can only predict genes associated with a specific disease at a time. Similarities among diseases are not used during the prediction. Meanwhile, most methods predict new disease genes based on known associations, making them unable to predict disease genes for diseases without known associated genes.In this study, a manifold learning-based method is proposed for predicting disease-gene associations by assuming that the geodesic distance between any disease and its associated genes should be shorter than that of other non-associated disease-gene pairs. The model maps the diseases and genes into a lower dimensional manifold based on the known disease-gene associations, disease similarity and gene similarity to predict new associations in terms of the geodesic distance between disease-gene pairs. In the 3-fold cross-validation experiments, our method achieves scores of 0.882 and 0.854 in terms of the area under of the receiver operating characteristic (ROC) curve (AUC) for diseases with more than one known associated genes and diseases with only one known associated gene, respectively. Further de novo studies on Lung Cancer and Bladder Cancer also show that our model is capable of identifying new disease-gene associations.
- Published
- 2019
- Full Text
- View/download PDF
7. Identifying Disease-Gene Associations With Graph-Regularized Manifold Learning.
- Author
-
Luo, Ping, Xiao, Qianghua, Wei, Pi-Jing, Liao, Bo, and Wu, Fang-Xiang
- Subjects
SIMILARITY (Geometry) ,GEODESIC distance ,BLADDER cancer ,MANIFOLDS (Mathematics) ,RECEIVER operating characteristic curves ,DISEASE complications - Abstract
Complex diseases are known to be associated with disease genes. Uncovering disease-gene associations is critical for diagnosis, treatment, and prevention of diseases. Computational algorithms which effectively predict candidate disease-gene associations prior to experimental proof can greatly reduce the associated cost and time. Most existing methods are disease-specific which can only predict genes associated with a specific disease at a time. Similarities among diseases are not used during the prediction. Meanwhile, most methods predict new disease genes based on known associations, making them unable to predict disease genes for diseases without known associated genes.In this study, a manifold learning-based method is proposed for predicting disease-gene associations by assuming that the geodesic distance between any disease and its associated genes should be shorter than that of other non-associated disease-gene pairs. The model maps the diseases and genes into a lower dimensional manifold based on the known disease-gene associations, disease similarity and gene similarity to predict new associations in terms of the geodesic distance between disease-gene pairs. In the 3-fold cross-validation experiments, our method achieves scores of 0.882 and 0.854 in terms of the area under of the receiver operating characteristic (ROC) curve (AUC) for diseases with more than one known associated genes and diseases with only one known associated gene, respectively. Further de novo studies on Lung Cancer and Bladder Cancer also show that our model is capable of identifying new disease-gene associations. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
8. Ichthyosis, psoriasiform dermatitis, and recurrent fungal infections in patients with biallelic mutations in PERP
- Author
-
F Khosravi-Bachehmir, Sirous Zeinali, Amir Hossein Saeidian, J S Park, Kambiz Kamyab-Hesari, Zahra Saffarian, Zahra Safaei Naraghi, Sadegh Khodavaisy, Jouni Uitto, Hassan Vahidnezhad, Hamidreza Mahmoudi, and Leila Youssefian
- Subjects
medicine.medical_specialty ,business.industry ,Ichthyosis ,Nonsense mutation ,Eczema ,Membrane Proteins ,Dermatology ,Iran ,Disease gene identification ,medicine.disease ,Pedigree ,Infectious Diseases ,Palmoplantar keratoderma ,Mycoses ,Psoriasis ,Mutation ,Humans ,Medicine ,Genes, Tumor Suppressor ,business ,Keratoderma ,Psoriasiform Dermatitis ,Guttate psoriasis - Abstract
BACKGROUND Germline autosomal dominant and autosomal recessive mutations in PERP encoding p53 effector related to PMP-22 (PERP), a component of epidermal desmosomes, have been associated with a spectrum of keratodermas. Monoallelic nonsense mutations cause Olmsted syndrome with severe periorificial and palmoplantar keratoderma (PPK). Biallelic recessive frameshift and missense mutations are associated with milder forms of the disease, including generalized erythrokeratoderma and PPK. OBJECTIVES To add new insights into the genotype-phenotype correlations as a consequence of PERP mutations and to provide a comprehensive review of the literature. METHODS Among 26 previously unresolved families within a cohort of 180 extended Iranian families with syndromic or nonsyndromic ichthyosis, two families with shared clinical features were examined by whole-exome sequencing (WES) and genome-wide homozygosity mapping (HM). Mycological and dermatopathological studies were performed to further characterize their atypical phenotypic presentations. RESULTS In two unrelated multiplex consanguineous families affected by ichthyosis, two novel biallelic PERP variants, NM_022121.5, c.89T>C, p.Leu30Pro and c.466G>C, p.Gly156Arg, located inside of genomic homozygosity regions of the probands were detected. Interestingly, some patients had areas of scaly psoriasiform plaques on a background of generalized ichthyosis that appeared during active cutaneous fungal infections. Mycological examinations of these lesions revealed infections caused by Candida albicans, Epidermophyton floccosum, or Trichophyton rubrum. Histopathology of the psoriasiform lesions shared some features with psoriasis, which when combined with clinical presentation, led to incorrect diagnoses of guttate psoriasis or pustular psoriasis. CONCLUSIONS PERP variants in ichthyosis patients can confer susceptibility to recalcitrant cutaneous fungal infections. Additionally, patients with episodic psoriasiform dermatitis in the setting of keratoderma should be considered for PERP genotyping and cutaneous fungal examinations.
- Published
- 2021
- Full Text
- View/download PDF
9. Novel variants in the LRP4 underlying Cenani-Lenz Syndactyly syndrome
- Author
-
Shabir Hussain, Muhammad Bilal, Shifeng Xue, Wasim Ahmad, Amir Hussain, Hamid Ali, Imran Ullah, Abdullah, Sanaullah Abbasi, Hammal Khan, Shoaib Nawaz, and Angie En Qi Chong
- Subjects
Genetics ,Sanger sequencing ,Mutant ,Wnt signaling pathway ,Biology ,Disease gene identification ,medicine.disease ,symbols.namesake ,medicine ,symbols ,Missense mutation ,Syndactyly ,Gene ,Genetics (clinical) ,Exome sequencing - Abstract
Cenani-Lenz syndrome (CLS) is a rare autosomal-recessive congenital disorder affecting development of distal limbs. It is characterized mainly by syndactyly and/or oligodactyly, renal anomalies, and characteristic facial features. Mutations in the LRP4 gene, located on human chromosome 11p11.2-q13.1, causes the CLS. The gene LRP4 encodes a low-density lipoprotein receptor-related protein-4, which mediates SOST-dependent inhibition of bone formation and Wnt signaling. In the study, presented here, three families of Pakistani origin, segregating CLS in the autosomal recessive manner were clinically and genetically characterized. In two families (A and B), microsatellite-based homozygosity mapping followed by Sanger sequencing identified a novel homozygous missense variant [NM_002334.3: c.295G>C; p.(Asp99His)] in the LRP4 gene. In the third family C, exome sequencing revealed a second novel homozygous missense variant [NM_002334.3: c.1633C>T; p.(Arg545Trp)] in the same gene. To determine the functional relevance of these variants, we tested their ability to inhibit canonical WNT signaling in a luciferase assay. Wild type LRP4 was able to inhibit LRP6-dependent WNT signaling robustly. The two mutants p.(Asp99His) and p.(Arg545Trp) inhibited WNT signaling less effectively, suggesting they reduced LRP4 function.
- Published
- 2021
- Full Text
- View/download PDF
10. A novel protein truncating mutation in L2HGDH causes L-2-hydroxyglutaric aciduria in a consanguineous Pakistani family
- Author
-
Christian Enzinger, Jasmin Blatterer, Christian Windpassinger, Saadullah Khan, Klaus Wagner, Safeer Ahmad, Muhammad Zeeshan Ali, Muzammil Ahmad Khan, Erwin Petek, Syed Khizar Shah, Sundas Taj, Beatrice A. Brugger, and Muhammad Muzammal
- Subjects
Male ,Intellectual disability ,Biochemistry ,Frameshift mutation ,Cellular and Molecular Neuroscience ,symbols.namesake ,Consanguinity ,medicine ,Humans ,Pakistan ,L2HGDH ,Exome ,Exome sequencing ,Genetics ,Sanger sequencing ,Cerebellar ataxia ,business.industry ,Macrocephaly ,Whole exome sequencing ,Leukoaraiosis ,N-terminal frameshift mutation ,Brain Diseases, Metabolic, Inborn ,Disease gene identification ,Alcohol Oxidoreductases ,L-2-hydroxyglutaric aciduria ,Mutation ,symbols ,Original Article ,Neurology (clinical) ,medicine.symptom ,business - Abstract
Background L-2-hydroxyglutaric aciduria (L2HGA) is a rare neurometabolic disorder that occurs due to accumulation of L-2-hydroxyglutaric acid in the cerebrospinal fluid (CSF), plasma and urine. The clinical manifestation of L2HGA includes intellectual disability, cerebellar ataxia, epilepsy, speech problems and macrocephaly. Methods In the present study, we ascertained a multigenerational consanguineous Pakistani family with 5 affected individuals. Clinical studies were performed through biochemical tests and brain CT scan. Locus mapping was carried out through genome-wide SNP genotyping, whole exome sequencing and Sanger sequencing. For in silico studies protein structural modeling and docking was done using I-TASSER, Cluspro and AutoDock VINA tools. Results Affected individuals presented with cognitive impairment, gait disturbance, speech difficulties and psychomotor delay. Radiologic analysis of a male patient revealed leukoaraiosis with hypoattenuation of cerebral white matter, suggestive of hypomyelination. Homozygosity mapping in this family revealed a linkage region on chromosome 14 between markers rs2039791 and rs781354. Subsequent whole exome analysis identified a novel frameshift mutation NM_024884.3:c.180delG, p.(Ala62Profs*24) in the second exon of L2HGDH. Sanger sequencing confirmed segregation of this mutation with the disease phenotype. The identification of the most N-terminal loss of function mutation published thus far further expands the mutational spectrum of L2HGDH.
- Published
- 2021
11. NDUFS6 mutations are a novel cause of lethal neonatal mitochondrial complex I deficiency
- Author
-
Edwin P. Kirk, Robert W. Taylor, Canny Sugiana, Denise M. Kirby, Avihu Boneh, Hans Henrik M. Dahl, Renato Salemi, Katrina M. Bell, Akira Ohtake, David R. Thorburn, Michael T. Ryan, and Lee Parry
- Subjects
Adult ,Male ,Mitochondrial DNA ,Adolescent ,Respiratory chain ,Biology ,medicine.disease_cause ,Models, Biological ,DNA, Mitochondrial ,Article ,Oxidative Phosphorylation ,Cell Line ,Cell Fusion ,medicine ,Humans ,Age of Onset ,QH426 ,Gene ,Genetics ,NDUFS6 ,Mutation ,Electron Transport Complex I ,Models, Genetic ,Point mutation ,Genetic Complementation Test ,NADH Dehydrogenase ,General Medicine ,Disease gene identification ,Molecular biology ,Mitochondria ,Pedigree ,Complementation ,Child, Preschool ,Lactates ,Commentary ,Female - Abstract
Complex I deficiency, the most common respiratory chain defect, is genetically heterogeneous: mutations in 8 nuclear and 7 mitochondrial DNA genes encoding complex I subunits have been described. However, these genes account for disease in only a minority of complex I–deficient patients. We investigated whether there may be an unknown common gene by performing functional complementation analysis of cell lines from 10 unrelated patients. Two of the patients were found to have mitochondrial DNA mutations. The other 8 represented 7 different (nuclear) complementation groups, all but 1 of which showed abnormalities of complex I assembly. It is thus unlikely that any one unknown gene accounts for a large proportion of complex I cases. The 2 patients sharing a nuclear complementation group had a similar abnormal complex I assembly profile and were studied further by homozygosity mapping, chromosome transfers, and microarray expression analysis. NDUFS6, a complex I subunit gene not previously associated with complex I deficiency, was grossly underexpressed in the 2 patient cell lines. Both patients had homozygous mutations in this gene, one causing a splicing abnormality and the other a large deletion. This integrated approach to gene identification offers promise for identifying other unknown causes of respiratory chain disorders.
- Published
- 2023
- Full Text
- View/download PDF
12. Autosomal‐dominant myopia associated to a novel <italic>P4HA2</italic> missense variant and defective collagen hydroxylation.
- Author
-
Gianfrancesco, F., Napolitano, F., Esposito, T., Lombardi, L., Farina, O., Di Iorio, G., Sampaolo, S., Melone, M. A. B., Di Iorio, V., Testa, F., Simonelli, F., Tirozzi, A., and Reccia, M. G.
- Subjects
- *
HYDROXYLATION , *COLLAGEN , *GENOTYPES , *PHENOTYPES , *MYOPIA - Abstract
We recently described a complex multisystem syndrome in which mild‐moderate myopia segregated as an independent trait. A plethora of genes has been related to sporadic and familial myopia. More recently, in Chinese patients severe myopia (MYP25, OMIM:617238) has been linked to mutations in
P4HA2 gene. Seven family members complaining of reduced distance vision especially at dusk underwent complete ophthalmological examination. Whole‐exome sequencing was performed to identify the gene responsible for myopia in the pedigree. Moderate myopia was diagnosed in the family which was associated to the novel missense variant c.1147A > G p.(Lys383Glu) in the prolyl 4‐hydroxylase,alpha‐polypeptide 2 (P4HA2 ) gene, which catalyzes the formation of 4‐hydroxyproline residues in the collagen strands. In vitro studies demonstratedP4HA2 mRNA and protein reduced expression level as well as decreased collagen hydroxylation and deposition in mutated fibroblast primary cultures compared to healthy cell lines. This study suggests thatP4HA2 mutations may lead to myopic axial elongation of eyeball as a consequence of quantitative and structural alterations of collagen. This is the first confirmatory study which associates a novel dominant missense variant inP4HA2 with myopia in Caucasian patients. Further studies in larger cohorts are advisable to fully clarify genotype‐phenotype correlations. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
13. A frameshift insertion in FA2H causes a recessively inherited form of ichthyosis congenita in Chianina cattle
- Author
-
Joana G. P. Jacinto, Cord Drögemüller, Irene M. Häfliger, Inês M. B. Veiga, Anna Letko, Arcangelo Gentile, Jacinto J.G.P., Hafliger I.M., Veiga I.M.B., Letko A., Gentile A., and Drogemuller C.
- Subjects
Candidate gene ,Genodermatose ,Hyperkeratosis ,Population ,Cattle Diseases ,610 Medicine & health ,Biology ,Mixed Function Oxygenases ,Frameshift mutation ,Urolithiasis ,Chromosome 18 ,Genodermatoses ,Genetics ,medicine ,Animals ,Genetic Predisposition to Disease ,Allele ,Frameshift Mutation ,education ,Molecular Biology ,Skin ,Sphingolipids ,education.field_of_study ,Genome ,Whole Genome Sequencing ,630 Agriculture ,Ichthyosis ,Precision medicine ,Bovine ,Fatty acid 2-hydroxylase ,General Medicine ,medicine.disease ,Disease gene identification ,590 Animals (Zoology) ,570 Life sciences ,biology ,Original Article ,Cattle ,Ichthyosis, Lamellar - Abstract
The aim of this study was to characterize the phenotype and to identify the genetic etiology of a syndromic form of ichthyosis congenita (IC) observed in Italian Chianina cattle and to estimate the prevalence of the deleterious allele in the population. Sporadic occurrence of different forms of ichthyosis including IC have been previously reported in cattle. However, so far, no causative genetic variant has been found for bovine IC. Nine affected cattle presenting congenital xerosis, hyperkeratosis and scaling of the skin as well as urolithiasis and cystitis associated with retarded growth were examined. Skin histopathology revealed a severe, diffuse orthokeratotic hyperkeratosis with mild to moderate epidermal hyperplasia. The pedigree records indicated a monogenic recessive trait. Homozygosity mapping and whole-genome sequencing allowed the identification of a homozygous frameshift 1 bp insertion in the FA2H gene (c.9dupC; p.Ala4ArgfsTer142) located in a 1.92 Mb shared identical-by-descent region on chromosome 18 present in all cases, while the parents were heterozygous as expected for obligate carriers. These findings enable the selection against this sub-lethal allele showing an estimated frequency of ~ 7.5% in Chianina top sires. A sporadic incidence of mild clinical signs in the skin of heterozygous carriers was observed. So far, pathogenic variants affecting the encoded fatty acid 2-hydroxylase catalyzing the synthesis of 2-hydroxysphingolipids have been associated with myelin disorders. In conclusion, this study represents the first report of an FA2H-related autosomal recessive inherited skin disorder in a mammalian species and adds FA2H to the list of candidate genes for ichthyosis in humans and animals. Furthermore, this study provides a DNA-based diagnostic test that enables selection against the identified pathogenic variant in the Chianina cattle population. However, functional studies are needed to better understand the expression of FA2H in IC-affected Chianina cattle.
- Published
- 2021
- Full Text
- View/download PDF
14. Loss of Function Variants in the XPC Causes Severe Xeroderma Pigmentosum in Three Large Consanguineous Families
- Author
-
Wasim Ahmad, Hammal Khan, Farooq Ahmad, Ubaid Ullah, Bushra Khan, Asmat Ullah, Gul Saeed Ahmad, Umm-e Kalsoom, Muhammad Ansar, Kanza Farrakh, and Warda Nawal
- Subjects
Sanger sequencing ,Genetics ,Xeroderma Pigmentosum ,Xeroderma pigmentosum ,Genetic heterogeneity ,Homozygote ,Chromosome ,Biology ,medicine.disease ,Disease gene identification ,DNA-Binding Proteins ,Consanguinity ,symbols.namesake ,Mutation ,Pediatrics, Perinatology and Child Health ,symbols ,medicine ,Humans ,Microsatellite ,Gene ,Nucleotide excision repair - Abstract
Background Xeroderma pigmentosum (XP) is a rare recessively inherited disorder that presents clinical and genetic heterogeneity. Mutations in eight genes, of which seven are involved in nucleotide excision repair (NER) pathway have been reported to cause the XP. Methods and Results Three large consanguineous families of Pakistani origin displaying typical clinical hallmarks of XP were evaluated at clinical and molecular level. Homozygosity mapping using microsatellite markers established linkage of the families to XPC gene on chromosome 3p25.1. Sanger sequencing of the XPC gene identified a novel homozygous single bp deletion [NM_004628.5; c.1934del; p.(Pro645Leufs*5)] and two previously reported mutations that included a nonsense [c.1243 C>T; p.(Arg415*)] and a splice acceptor site (c.2251–1 G>C), all segregating with the disease phenotypes in the families. Conclusion This report has extended the spectrum of mutations in the XPC gene and will also facilitate in diagnosis of XP and counselling of families inheriting it, which is the only inevitable tool for preventing the disease occurrence in future generations.
- Published
- 2021
- Full Text
- View/download PDF
15. A discarded synonymous variant in NPHP3 explains nephronophthisis and congenital hepatic fibrosis in several families
- Author
-
Ian J. Wilson, Mohammed S. Al Riyami, Khawla A Rahim, Eric Olinger, Elisa Molinari, Laura Powell, Miguel Barroso-Gil, Abdulrahman Al-Hussaini, Naif A.M. Almontashiri, Shirlee Shril, John A. Sayer, Mohamed H Al-Hamed, Friedhelm Hildebrandt, Intisar Al Alawi, Colin G. Miles, Isa Al Salmi, and Eissa Ali Faqeih
- Subjects
Liver Cirrhosis ,Genetics ,Polycystic Kidney Diseases ,Candidate gene ,Homozygote ,Genetic Diseases, Inborn ,Kinesins ,Biology ,Disease gene identification ,medicine.disease ,Article ,Frameshift mutation ,Ciliopathy ,Nephronophthisis ,Exome Sequencing ,medicine ,Humans ,Congenital hepatic fibrosis ,Allele ,Child ,Genetics (clinical) ,Exome sequencing - Abstract
Half of patients with a ciliopathy syndrome remain unsolved after initial analysis of whole exome sequencing (WES) data, highlighting the need for improved variant filtering and annotation. By candidate gene curation of WES data, combined with homozygosity mapping, we detected a homozygous predicted synonymous allele in NPHP3 in two children with hepatorenal fibrocystic disease from a consanguineous family. Analyses on patient-derived RNA shows activation of a cryptic mid-exon splice donor leading to frameshift. Remarkably, the same rare variant was detected in four additional families with hepatorenal disease from UK, US, and Saudi patient cohorts and in addition, another synonymous NPHP3 variant was identified in an unsolved case from the Genomics England 100,000 Genomes data set. We conclude that synonymous NPHP3 variants, not reported before and discarded by pathogenicity pipelines, solved several families with a ciliopathy syndrome. These findings prompt careful reassessment of synonymous variants, especially if they are rare and located in candidate genes.
- Published
- 2021
- Full Text
- View/download PDF
16. <scp> ANXA1 </scp> with Anti‐Inflammatory Properties Might Contribute to Parkinsonism
- Author
-
Shaghayegh Taghavi, Hossein Darvish, Safoura Zardadi, Elham Alehabib, Fatemeh Mohajerani, Luis J. Azcona, Abbas Tafakhori, Saghar Ghasemi Firouzabadi, and Coro Paisán-Ruiz
- Subjects
Annexins ,Neuroprotection ,Parkinsonian Disorders ,medicine ,Animals ,Humans ,SNP ,Zebrafish ,Inflammation ,Whole genome sequencing ,Genetics ,biology ,Microglia ,Siblings ,Parkinsonism ,Genetic Variation ,Middle Aged ,Disease gene identification ,biology.organism_classification ,medicine.disease ,Pedigree ,medicine.anatomical_structure ,Neurology ,Female ,Neurology (clinical) ,Annexin A1 - Abstract
We here describe the identification of a novel variant in the anti-inflammatory Annexin A1 protein likely to be the cause of disease in two siblings with autosomal recessive parkinsonism. The disease-segregating variant was ascertained through a combination of homozygosity mapping and whole genome sequencing and was shown to impair phagocytosis in zebrafish mutant embryos. The highly conserved variant, absent in healthy individuals and public SNP databases, affected a functional domain of the protein with neuroprotective properties. This study supports the hypothesis that damaged microglia might lead to impairments in the clearance of accumulated and aggregated proteins resulting in parkinsonism. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
17. Risk homozygous haplotype regions for autism identifies population-specific ten genes for numerous pathways
- Author
-
Nallur B. Ramachandra and Swati Agarwala
- Subjects
Candidate gene ,Recessive genes ,Autism ,Population ,Single-nucleotide polymorphism ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Quantitative trait locus ,Biology ,Homozygosity ,03 medical and health sciences ,0302 clinical medicine ,medicine ,SNP ,education ,030304 developmental biology ,Genetics ,0303 health sciences ,education.field_of_study ,General Neuroscience ,Haplotype ,Haplotype blocks ,Disease gene identification ,medicine.disease ,Psychiatry and Mental health ,Surgery ,Neurology (clinical) ,Pshychiatric Mental Health ,030217 neurology & neurosurgery ,Mutations ,RC321-571 - Abstract
Background Recessive homozygous haplotype (rHH) mapping is a reliable tool for identifying recessive genes by detecting homozygous segments of identical haplotype structures. These are shared at a higher frequency amongst probands compared to parental controls. Finding out such rHH blocks in autism subjects can help in deciphering the disorder etiology. Objectives The study aims to detect rHH segments of identical haplotype structure shared at a higher frequency in autism subjects than controls to identify recessive genes responsible for autism manifestation. Methods In the present study, 426 unrelated autism genotyped probands with 232 parents (116 trios) were obtained from Gene Expression Omnibus (GEO) Database. Homozygosity mapping analyses have been performed on the samples using standardized algorithms using the Affymetrix GeneChip® 500K SNP Nsp and Sty mapping arrays datasets. Results A total of 38 homozygous haplotype blocks were revealed across sample datasets. Upon downstream analysis, 10 autism genes were identified based on selected autism candidate genes criteria. Further, expressive Quantitative Trait Loci (QTL) analysis of SNPs revealed various binding sites for regulatory proteins BX3, FOS, BACH1, MYC, JUND, MAFK, POU2F2, RBBP5, RUNX3, and SMARCA4 impairing essential autism genes CEP290, KITLG, CHD8, and INS2. Pathways and processes such as adherens junction, dipeptidase activity, and platelet-derived growth factor—vital to autism manifestation were identified with varied protein-protein clustered interactions. Conclusion These findings bring various population clusters with significant rHH genes. It is suggestive of the existence of common but population-specific risk alleles in related autism subjects.
- Published
- 2021
18. History of the methodology of disease gene identification
- Author
-
Stylianos E. Antonarakis
- Subjects
0301 basic medicine ,Positional cloning ,Genetic Linkage ,Computational biology ,030105 genetics & heredity ,Biology ,History, 21st Century ,03 medical and health sciences ,Genetic linkage ,Genetic variation ,Databases, Genetic ,Genetics ,Humans ,Genetic Predisposition to Disease ,Gene ,Genetics (clinical) ,Sequence (medicine) ,gene identification ,Mendelian disorders ,In Memoriam ,Genetic Diseases, Inborn ,Genomics ,History, 20th Century ,Disease gene identification ,Phenotype ,genetic methods ,genomic variants ,030104 developmental biology ,Identification (biology) - Abstract
The past 45 years have witnessed a triumph in the discovery of genes and genetic variation that cause Mendelian disorders due to high impact variants. Important discoveries and organized projects have provided the necessary tools and infrastructure for the identification of gene defects leading to thousands of monogenic phenotypes. This endeavor can be divided in three phases in which different laboratory strategies were employed for the discovery of disease‐related genes: (i) the biochemical phase, (ii) the genetic linkage followed by positional cloning phase, and (iii) the sequence identification phase. However, much more work is needed to identify all the high impact genomic variation that substantially contributes to the phenotypic variation.
- Published
- 2021
19. Mutational analysis of CYP1B1 gene in Iranian pedigrees with glaucoma reveals known and novel mutations
- Author
-
Somayeh Kazeminasb, Elham Alehabib, Zahra Bahmanpour, Hossein Darvish, Minoo Atakhorrami, Babak Emamalizadeh, Ehsan Aghaei Moghadam, Yousef Daneshmandpour, Somayeh Alinaghi, and Azadeh Doozandeh
- Subjects
DNA Mutational Analysis ,Iran ,03 medical and health sciences ,Exon ,symbols.namesake ,0302 clinical medicine ,Genetic linkage ,Humans ,Missense mutation ,Medicine ,Gene ,Genetics ,Sanger sequencing ,business.industry ,Glaucoma ,Disease gene identification ,Phenotype ,Pedigree ,body regions ,Ophthalmology ,genomic DNA ,Latent TGF-beta Binding Proteins ,Cytochrome P-450 CYP1B1 ,Mutation ,030221 ophthalmology & optometry ,symbols ,business ,030217 neurology & neurosurgery - Abstract
Primary congenital glaucoma (PCG) (OMIM#231,300) can be caused by pathogenic sequence variations in CYP1B1, LTBP2, MYOC and PXDN genes. The purpose of this study was to investigate mutations in the CYP1B1 gene in families affected with primary congenital glaucoma (PCG) using linkage analysis and Sanger sequencing. A total number of four families with nine affected PCG patients during six months were included in this study. The mutations were identified by homozygosity mapping to find the linked loci and then direct sequencing of all coding exons, the exon–intron boundaries and the 5' untranslated region of CYP1B1 using genomic DNA obtained from affected family members and their parents. Moreover, bioinformatic tools were applied to study mutation effect on protein structure and function. A total of four mutations were identified, and three of these were novel. Two were missense mutations: One was truncating mutation, and the other was an in-frame deletion. Mutations in CYP1B1 could fully explain the PCG phenotype in all of the patients. Also, the bioinformatic study of the mutations showed the structure of the protein is affected, and it is well conserved among similar species. In this study, we identified 4 CYP1B1 mutations, 3 of which were novel. In silico analysis of identified mutations confirmed their molecular pathogenicity. A similar analysis will help understand the biological role of CYP1B1 and the effect of mutations on the regulatory and enzymatic functions of CYP1B1 that result in PCG. Not relevant
- Published
- 2021
- Full Text
- View/download PDF
20. Pathogenic STX3 variants affecting the retinal and intestinal transcripts cause an early-onset severe retinal dystrophy in microvillus inclusion disease subjects
- Author
-
Andreas R. Janecke, Lukas A. Huber, Roberto Adachi, Valeria Strauß, Anuradha Ganesh, Siham Al Sinani, Martha P. Schatz, Roger Janz, Badr AlSaleem, Martin Walter Laass, Majid Alfadhel, Sumanth Punuru, Elizabeth Sanchez, Xiaoqin Liu, Naveen Mittal, Fowzan S. Alkuraya, Ujwala S. Saboo, Rüdiger Adam, Sana Al Zuhaibi, Klaus Rohrschneider, Fathiya Al Murshedi, Thomas Müller, Ruth Heidelberger, Johanna C. Escher, Arne Viestenz, and Pediatrics
- Subjects
chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Mucolipidoses ,Enteropathy ,Intestinal Mucosa ,Genetics (clinical) ,Exome sequencing ,Original Investigation ,Genetics ,Aged, 80 and over ,Mice, Knockout ,0303 health sciences ,Microvilli ,Qa-SNARE Proteins ,Homozygote ,Eye Diseases, Hereditary ,Disease gene identification ,Phenotype ,medicine.anatomical_structure ,Knockout mouse ,Retinal Cone Photoreceptor Cells ,Female ,Autopsy ,Co-Repressor Proteins ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Malabsorption Syndromes ,SDG 3 - Good Health and Well-being ,Retinal Dystrophies ,Exome Sequencing ,medicine ,Animals ,Humans ,Sensory Rhodopsins ,RNA, Messenger ,Outer nuclear layer ,030304 developmental biology ,Aged ,Retinal ,medicine.disease ,Human genetics ,Alcohol Oxidoreductases ,chemistry ,Gene Expression Regulation ,sense organs ,030217 neurology & neurosurgery - Abstract
Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic—intestinal and retinal—disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.
- Published
- 2021
21. In-frame deletion in canine PITRM1 is associated with a severe early-onset epilepsy, mitochondrial dysfunction and neurodegeneration
- Author
-
Hannes Lohi, Anu Suomalainen, Marjo K. Hytönen, Kaspar Matiasek, Meharji Arumilli, Ileana B. Quintero, Enrico Baruffini, Tarja S. Jokinen, Jonas Donner, Geoffray Monteuuis, Marjukka Anttila, Cristina Dallabona, Laurence A. Bindoff, Christopher B. Jackson, Marco Rosati, Pernilla Syrjä, Riika Sarviaho, Medicum, Department of Medical and Clinical Genetics, Veterinary Biosciences, STEMM - Stem Cells and Metabolism Research Program, Veterinary Genetics, Department of Biochemistry and Developmental Biology, Veterinary Pathology and Parasitology, Antti Sukura / Principal Investigator, Helsinki One Health (HOH), Departments of Faculty of Veterinary Medicine, Equine and Small Animal Medicine, Institute for Molecular Medicine Finland, Hannes Tapani Lohi / Principal Investigator, Department of Neurosciences, Anu Wartiovaara / Principal Investigator, HUSLAB, Haartman Institute (-2014), and Biosciences
- Subjects
Male ,medicine.medical_specialty ,Respiratory chain ,Status epilepticus ,Saccharomyces cerevisiae ,Grey matter ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Dogs ,Oxygen Consumption ,Internal medicine ,Genetics ,medicine ,Animals ,Dog Diseases ,Genetics (clinical) ,030304 developmental biology ,Original Investigation ,0303 health sciences ,Mutation ,Amyloid beta-Peptides ,Neurodegeneration ,1184 Genetics, developmental biology, physiology ,Brain ,Metalloendopeptidases ,Neurodegenerative Diseases ,medicine.disease ,Disease gene identification ,3. Good health ,Mitochondria ,Pedigree ,medicine.anatomical_structure ,Endocrinology ,Spinocerebellar ataxia ,Female ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
We investigated the clinical, genetic, and pathological characteristics of a previously unknown severe juvenile brain disorder in several litters of Parson Russel Terriers. The disease started with epileptic seizures at 6 to 12 weeks of age and progressed rapidly to status epilepticus and death or euthanasia. Histopathological changes at autopsy were restricted to the brain. There was severe acute neuronal degeneration and necrosis diffusely affecting the grey matter throughout the brain with extensive intraneuronal mitochondrial crowding and accumulation of amyloid-β (Aβ). Combined homozygosity mapping and genome sequencing revealed an in-frame 6-bp deletion in the nuclear-encoded pitrilysin metallopeptidase 1 (PITRM1) encoding for a mitochondrial protease involved in mitochondrial targeting sequence processing and degradation. The 6-bp deletion results in the loss of two amino acid residues in the N-terminal part of PITRM1, potentially affecting protein folding and function. Assessment of the mitochondrial function in the affected brain tissue showed a significant deficiency in respiratory chain function. The functional consequences of the mutation were modeled in yeast and showed impaired growth in permissive conditions and an impaired respiration capacity. Loss-of-function variants in human PITRM1 result in a childhood-onset progressive amyloidotic neurological syndrome characterized by spinocerebellar ataxia with behavioral, psychiatric and cognitive abnormalities. Homozygous Pitrm1-knockout mice are embryonic lethal, while heterozygotes show a progressive, neurodegenerative phenotype characterized by impairment in motor coordination and Aβ deposits. Our study describes a novel early-onset PITRM1-related neurodegenerative canine brain disorder with mitochondrial dysfunction, Aβ accumulation, and lethal epilepsy. The findings highlight the essential role of PITRM1 in neuronal survival and strengthen the connection between mitochondrial dysfunction and neurodegeneration.
- Published
- 2021
22. Exome sequencing and functional studies in zebrafish identify WDR8 as the causative gene for isolated Microspherophakia in Indian families
- Author
-
Debanjan Dutta, Maheswara R Duvvari, Ankana Tiwari, Gowri J Murthy, Sautan Show, Upendra Nongthomba, Mohammad Iqbal Rather, M Madhangi, Nivedita Singh, Vishwanath Kumble Bhat, and Arun Kumar
- Subjects
Adult ,Male ,0301 basic medicine ,Mutant ,India ,Iris ,medicine.disease_cause ,Ectopia Lentis ,Corneal Diseases ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Ciliogenesis ,Exome Sequencing ,Genetics ,medicine ,Animals ,Humans ,Missense mutation ,Exome ,Child ,Molecular Biology ,Zebrafish ,Genetics (clinical) ,Exome sequencing ,Mutation ,biology ,Proteins ,Glaucoma ,General Medicine ,Disease gene identification ,medicine.disease ,biology.organism_classification ,Pedigree ,030104 developmental biology ,Microspherophakia ,Female ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Isolated Microspherophakia (MSP) is an autosomal recessive disorder characterized by a smaller than normal spherical lens. Till date, LTBP2 is the only gene shown to cause MSP. We used homozygosity mapping and whole-exome sequencing and identified a homozygous mutation, c.1148C > T (p.Pro383Leu), in the WDR8 (or WRAP73) gene in two Indian MSP families. In vitro experiments showed that the missense mutation renders the protein unstable. WDR8 is a centriolar protein that has important roles in centrosomal assembly, spindle pole formation and ciliogenesis. Co-immunoprecipitation experiments from HeLa cells indicated that the mutation interferes with the interaction of WDR8 with its binding partners. In zebrafish, both morpholino-mediated knockdown and CRISPR/Cas knockout of wdr8 resulted in decreased eye and lens size. The lack of wdr8 affected cell cycle progression in the retinal cells, causing a reduction in cell numbers in the retina and lens. The reduction in eye size and the cell cycle defects were rescued by exogenous expression of the human wild-type WDR8. However, the human mutant WDR8 (p.Pro383Leu) was unable to rescue the eye defects, indicating that the missense mutation abrogates WDR8 protein function. Thus, our zebrafish results suggested that WDR8 is the causative gene for MSP in these Indian families.
- Published
- 2021
- Full Text
- View/download PDF
23. Identification of Three Novel and One Known Mutation in the WFS1 Gene in Four Unrelated Turkish Families: The Role of Homozygosity Mapping in the Early Diagnosis
- Author
-
Khalid Hussain, Meliha Demiral, Edip Unal, Dogus Vuralli, Riza Taner Baran, Mehmet Nuri Ozbek, Ayşe Nurcan Cebeci, Gonul Buyukyilmaz, Sophia Tahir, Maha Sherif, Atilla Cayir, Busra Cavdarli, Huseyin Demirbilek, and Sofia A. Rahman
- Subjects
Wolfram syndrome ,Endocrinology, Diabetes and Metabolism ,wolfram syndrome ,Pedigree chart ,medicine.disease_cause ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,sensorineural deafness ,Endocrinology ,medicine ,optic atrophy ,Gene ,Exome sequencing ,Genetics ,Mutation ,lcsh:RC648-665 ,business.industry ,lcsh:RJ1-570 ,lcsh:Pediatrics ,medicine.disease ,Disease gene identification ,Stop codon ,Chromosome 4 ,diabetes insipidus ,Pediatrics, Perinatology and Child Health ,diabetes mellitus ,business ,wfs1 - Abstract
Objective Bi-allelic mutations in wolframin gene (WFS1) cause Wolfram syndrome 1 (WS1 or DIDMOAD) characterized with non-autoimmune diabetes mellitus (DM), optic atrophy (OA), diabetes insipidus (DI), sensorineural deafness (SND), urinary tract abnormalities, and neuropsychiatric disorders. We evaluated patients presented with an incomplete phenotype of WS1 using homozygosity mapping and subsequent whole-exome sequencing. Patients and methods Four unrelated consanguineous Turkish families (7 affected children), and their unaffected parents and siblings were evaluated. Homozygosity mapping (HZM) was performed, followed by whole-exome sequencing of WFS1. Mutations were classified according to results of "in silico" analyses, protein prediction, and functional consequences. Results Homozygosity mapping confirmed shared homozygous regions on chromosome 4 (chr4p16.1) between the affected individuals, and absent from their unaffected siblings. Exome sequencing identified three novel (c.1215T>A, c.554G>A, c.1525_1540dup) and one known (c. 1522_1523delTA) mutations in WFS1 gene. All mutations were predicted to cause stop codon leading to early termination of protein synthesis and complete loss-of-function. All patients were found to be homozygous for the change, with parents and other unaffected siblings being carriers. Conclusion Our study expands the mutation spectrum of WSF1 mutations with three novel mutations. Homozygosity mapping may provide enrichment for molecular genetic analysis and early diagnosis of WS1 patients with incomplete phenotype, particularly in consanguineous pedigrees.
- Published
- 2021
24. Mutations in PRDM15 Are a Novel Cause of Galloway-Mowat Syndrome
- Author
-
Ana C. Onuchic-Whitford, Florian Buerger, Slim Mzoughi, Denny Schanze, Beate Ermisch-Omran, Andreas R. Janecke, Susanne J. Kühl, Sven Schumann, Amy Kolb, Anja Werberger, Svjetlana Lovric, Shasha Shi, Verena Klämbt, Neveen A. Soliman, Youying Mao, Tilman Jobst-Schwan, Alma Kuechler, Ronen Schneider, Dagmar Wieczorek, Weizhen Tan, Jan Kadlec, Nina Mann, Franziska Kause, Amar J. Majmundar, Shrikant Mane, Kristina Holton, Ernesto Guccione, Thomas M. Kitzler, Martin Zenker, Amelie T. van der Ven, Makiko Nakayama, Thomas Lennert, Jia Rao, Oliver Gross, Michael J. Schmeisser, Eva Mildenberger, Martin Skalej, Daniela A. Braun, Shirlee Shril, Ernestine Treimer, Richard P. Lifton, Friedhelm Hildebrandt, and Michael Kühl
- Subjects
0301 basic medicine ,Genetics ,Kidney ,Medizin ,General Medicine ,Biology ,Disease gene identification ,medicine.disease ,Phenotype ,3. Good health ,Nephropathy ,Galloway Mowat syndrome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Nephrology ,Genetic linkage ,medicine ,Gene ,Nephrotic syndrome ,030217 neurology & neurosurgery - Abstract
Background Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. Methods Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. Results Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. Conclusions Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.
- Published
- 2021
- Full Text
- View/download PDF
25. Significance and Functional Similarity for Identification of Disease Genes.
- Author
-
Maji, Pradipta and Shah, Ekta
- Abstract
One of the most significant research issues in functional genomics is insilico identification of disease related genes. In this regard, the paper presents a new gene selection algorithm, termed as SiFS, for identification of disease genes. It integrates the information obtained from interaction network of proteins and gene expression profiles. The proposed SiFS algorithm culls out a subset of genes from microarray data as disease genes by maximizing both significance and functional similarity of the selected gene subset. Based on the gene expression profiles, the significance of a gene with respect to another gene is computed using mutual information. On the other hand, a new measure of similarity is introduced to compute the functional similarity between two genes. Information derived from the protein-protein interaction network forms the basis of the proposed SiFS algorithm. The performance of the proposed gene selection algorithm and new similarity measure, is compared with that of other related methods and similarity measures, using several cancer microarray data sets. [ABSTRACT FROM PUBLISHER]
- Published
- 2017
- Full Text
- View/download PDF
26. P.arg102ser is a common Pde6? mutation causing autosomal recessive retinitis pigmentosa in Pakistani families
- Author
-
null Anosha Ali Khan, null Yar Muhammad Waryah, null Muhammad Iqbal, null Hafiz Muhammad Azhar Baig, null Muhammad Rafique, null Ali Muhammad Waryah, and null Admin
- Subjects
Adult ,Adolescent ,Genetic counseling ,Genes, Recessive ,Pedigree chart ,Consanguinity ,Young Adult ,Genetic linkage ,Retinitis pigmentosa ,medicine ,Humans ,Pakistan ,Family history ,Child ,Eye Proteins ,Aged ,Aged, 80 and over ,Genetics ,Cyclic Nucleotide Phosphodiesterases, Type 6 ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Disease gene identification ,Child, Preschool ,Mutation ,Mutation (genetic algorithm) ,Microsatellite ,business ,Retinitis Pigmentosa - Abstract
Aim: To explore the genetic cause of autosomal recessive retinitis pigmentosa in consanguineous families. Methods: The multi-centre study was conducted from July 2015 to June 2018 at Liaquat University of Medical and health Sciences, Jamshoro, the University of Sindh, Jamshoro, and Islamia University, Bahawalpur, Pakistan, and comprised families affected with non-syndromic autosomal recessive retinitis pigmentosa. Ophthalmological investigations were done to assess the fundus of the patients and the status of the disease. Pedigrees were drawn and family histories were recorded to find out the mode of inheritance. A 10cc sample of whole blood was obtained from each participant and deoxyribonucleic acid was extracted. Homozygosity mapping was performed using three short tandem repeat polymorphisms closely linked to phosphodiesterase 6A gene, and the linked families were Sanger-sequenced for identification of the mutation. Bioinformatic tools were used to design amplification refractory mutation system assay and to assess the protein structure and pathogenic effects of the mutation. Results: In the 80 consanguineous families, there were 464 individuals, and, of them, 236(51%) were affected with their age ranging between 4 and 80 years. Family history and pedigree drawings revealed autosomal recessive retinitis pigmentosa with early childhood onset. Linkage analysis indicated the homozygosity in 6(7.5%) families. Sanger-sequencing revealed a common mutation c.304C>A (p.Arg102Ser); segregating with the disease in the linked families. Conclusion: The findings may offer effective genetic counselling and minimise disease penetration in consanguineous families. Key Words: PDE6a mutations, Retinitis pigmentosa, Pakistan, ARMS assay.
- Published
- 2020
- Full Text
- View/download PDF
27. <scp> WDR34 </scp> , a candidate gene for non‐syndromic rod‐cone dystrophy
- Author
-
Wassila Carpentier, Thierry Léveillard, Christine Lonjou, Jean-Paul Saraiva, Christel Condroyer, Saddek Mohand-Said, Shomi S. Bhattacharya, Hélène Dollfus, Maria Solaguren-Beascoa, Vanessa Démontant, Lisa Emmenegger, Marie-Elise Lancelot, Elise Orhan, Kinga M. Bujakowska, Christelle Michiels, Marion Neuillé, José-Alain Sahel, Isabelle Audo, Cécile Méjécase, Eric A. Pierce, Mélanie Letexier, Christina Zeitz, Aline Antonio, Ministère de l’Enseignement supérieur et de la Recherche (France), Fondation de France, Foundation Fighting Blindness, Fondation Voir et Entendre, Agence Nationale de la Recherche (France), National Eye Institute (US), Région Ile-de-France, and Association Française contre les Myopathies
- Subjects
Adult ,Male ,0301 basic medicine ,Candidate gene ,WD40 Repeats ,genetic structures ,WDR34 ,Whole-exomesequencing ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Retinitis pigmentosa ,Genetics ,Rod-cone dystrophy ,medicine ,Humans ,Genetic Association Studies ,Genetics (clinical) ,Exome sequencing ,Non-syndromic rod-cone dystrophy ,Mutation ,Dystrophy ,Disease gene identification ,medicine.disease ,Pedigree ,KIAA2026 ,Ciliopathy ,030104 developmental biology ,sense organs ,Carrier Proteins ,Cone-Rod Dystrophies - Abstract
Rod-cone dystrophy (RCD), also called retinitis pigmentosa, is characterized by rod followed by cone photoreceptor degeneration, leading to gradual visual loss. Mutations in over 65 genes have been associated with non-syndromic RCD explaining 60% to 70% of cases, with novel gene defects possibly accounting for the unsolved cases. Homozygosity mapping and whole-exome sequencing applied to a case of autosomal recessive non-syndromic RCD from a consanguineous union identified a homozygous variant in WDR34. Mutations in WDR34 have been previously associated with severe ciliopathy syndromes possibly associated with a retinal dystrophy. This is the first report of a homozygous mutation in WDR34 associated with non-syndromic RCD., Doctoral funding from the Ministère de l'Enseignement Supérieur et de la Recherche; Europe exchange 2018 Erasmus; European Reintegration Grant, Grant/Award Number: PERG04-GA-2008-231125; Fondation de France-Berthe Fouassier; Foundation Fighting Blindness, Grant/Award Number: Grant # CD-CL-0808-0466-CHNO CIC503 recogn; Foundation Voir et Entendre; French Agence Nationale de la Recherche, Grant/Award Numbers: IHU FOReSIGHT: ANR-18-IAHU-0001, LIFESENSES: ANR-10-LABX-65; National Eye Institute [R01EY012910 (EAP), R01EY026904 (KMB/EAP) and P30EY014104 (MEEI core support)], the Foundation Fighting
- Published
- 2020
- Full Text
- View/download PDF
28. A Recessively Inherited Risk Locus on Chromosome 13q22-31 Conferring Susceptibility to Schizophrenia
- Author
-
Nancy Kiwan, Peter W.R. Woodruff, Colin A. Johnson, Anneka Tomlinson, Steven J. Clapcote, Hassen Al-Amin, Iain D. Wilkinson, Sophia Ahmed, Joanna C. Neill, Jamshid Nazari, Ahmed H Al-Amri, Chris F. Inglehearn, Rifka S Chamali, Manir Ali, Tariq Mahmood, Mohammed E El-Asrag, Juliette Randerson Moor, Suhaila Ghuloum, Shabana Khan, Alastair G. Cardno, Hamid A. Alhaj, and James A. Poulter
- Subjects
Male ,AcademicSubjects/MED00810 ,Endophenotypes ,Genes, Recessive ,Locus (genetics) ,Biology ,DNA sequencing ,Consanguinity ,03 medical and health sciences ,0302 clinical medicine ,Humans ,consanguineous/endophenotype/homozygosity/ chromosome 13q/risk haplotype ,Genetic Predisposition to Disease ,Gene ,Exome ,030304 developmental biology ,Genetics ,0303 health sciences ,Chromosomes, Human, Pair 13 ,Haplotype ,Disease gene identification ,Pedigree ,Minor allele frequency ,Psychiatry and Mental health ,Psychotic Disorders ,Genetic Loci ,Endophenotype ,Schizophrenia ,Female ,030217 neurology & neurosurgery ,Regular Articles - Abstract
We report a consanguineous family in which schizophrenia segregates in a manner consistent with recessive inheritance of a rare, partial-penetrance susceptibility allele. From 4 marriages between 2 sets of siblings who are half first cousins, 6 offspring have diagnoses of psychotic disorder. Homozygosity mapping revealed a 6.1-Mb homozygous region on chromosome 13q22.2-31.1 shared by all affected individuals, containing 13 protein-coding genes. Microsatellite analysis confirmed homozygosity for the affected haplotype in 12 further apparently unaffected members of the family. Psychiatric reports suggested an endophenotype of milder psychiatric illness in 4 of these individuals. Exome and genome sequencing revealed no potentially pathogenic coding or structural variants within the risk haplotype. Filtering for noncoding variants with a minor allele frequency of
- Published
- 2020
- Full Text
- View/download PDF
29. Cancer Diagnosis and Disease Gene Identification via Statistical Machine Learning
- Author
-
Li Juntao, Chang Mingming, and Chen Liuyuan
- Subjects
Computational Mathematics ,Computer science ,Genetics ,medicine ,Cancer ,Computational biology ,medicine.disease ,Disease gene identification ,Molecular Biology ,Biochemistry - Abstract
Diagnosing cancer and identifying the disease gene by using DNA microarray gene expression data are the hot topics in current bioinformatics. This paper is devoted to the latest development in cancer diagnosis and gene selection via statistical machine learning. A support vector machine is firstly introduced for the binary cancer diagnosis. Then, 1-norm support vector machine, doubly regularized support vector machine, adaptive huberized support vector machine and other extensions are presented to improve the performance of gene selection. Lasso, elastic net, partly adaptive elastic net, group lasso, sparse group lasso, adaptive sparse group lasso and other sparse regression methods are also introduced for performing simultaneous binary cancer classification and gene selection. In addition to introducing three strategies for reducing multiclass to binary, methods of directly considering all classes of data in a learning model (multi_class support vector, sparse multinomial regression, adaptive multinomial regression and so on) are presented for performing multiple cancer diagnosis. Limitations and promising directions are also discussed.
- Published
- 2020
- Full Text
- View/download PDF
30. A 50-kb deletion disrupting the RSPO2 gene is associated with tetradysmelia in Holstein Friesian cattle
- Author
-
Christoph Schulze, Doreen Becker, Rosemarie Weikard, Peter Wohlsein, and Christa Kühn
- Subjects
lcsh:QH426-470 ,[SDV]Life Sciences [q-bio] ,Limb Deformities, Congenital ,Cattle Diseases ,Single-nucleotide polymorphism ,Biology ,medicine.disease_cause ,Chromosomes ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Coding region ,Gene ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,lcsh:SF1-1100 ,2. Zero hunger ,0303 health sciences ,Mutation ,Sire ,Haplotype ,General Medicine ,Disease gene identification ,lcsh:Genetics ,Chromosomal region ,Cattle ,Animal Science and Zoology ,lcsh:Animal culture ,Thrombospondins ,Gene Deletion ,030217 neurology & neurosurgery ,Research Article - Abstract
BackgroundTetradysmelia is a rare genetic disorder that is characterized by an extremely severe reduction of all limb parts distal of the scapula and pelvic girdle. We studied a Holstein Friesian backcross family with 24 offspring, among which six calves displayed autosomal recessive tetradysmelia. In order to identify the genetic basis of the disorder, we genotyped three affected calves, five dams and nine unaffected siblings using a Bovine Illumina 50 k BeadChip and sequenced the whole genome of the sire.ResultsPathological examination of four tetradysmelia cases revealed a uniform and severe dysmelia of all limbs. Applying a homozygosity mapping approach, we identified a homozygous region of 10.54 Mb on chromosome 14 (Bos taurusBTA14). Only calves that were diagnosed with tetradysmelia shared a distinct homozygous haplotype for this region. We sequenced the whole genome of the cases’ sire and searched for heterozygous single nucleotide polymorphisms (SNPs) and small variants on BTA14 that were uniquely present in the sire and absent from 3102 control whole-genome sequences of the 1000 Bull Genomes Project, but none were identified in the 10.54-Mb candidate region on BTA14. Therefore, we subsequently performed a more comprehensive analysis by also considering structural variants and detected a 50-kb deletion in the targeted chromosomal region that was in the heterozygous state in the cases’ sire. Using PCR, we confirmed that this detected deletion segregated perfectly within the family with tetradysmelia. The deletion spanned three exons of the bovineR-spondin 2(RSPO2) gene, which encode three domains of the respective protein. R-spondin 2 is a secreted ligand of leucine-rich repeats containing G protein-coupled receptors that enhance Wnt signalling and is involved in a broad range of developmental processes during embryogenesis.ConclusionsWe identified a 50-kb deletion on BTA14 that disrupts the coding sequence of theRSPO2gene and is associated with bovine tetradysmelia. To our knowledge, this is the first reported candidate causal mutation for tetradysmelia in a large animal model. Since signalling pathways involved in limb development are conserved across species, the observed inherited defect may serve as a model to further elucidate fundamental pathways of limb development.
- Published
- 2020
- Full Text
- View/download PDF
31. Homozygosity mapping and next generation sequencing for the genetic diagnosis of hereditary ataxia and spastic paraplegia in consanguineous families
- Author
-
Juan Du, Hong Jiang, Lu Shen, Xinxiang Yan, Bin Jiao, Yingying Luo, Beisha Tang, Xinxin Liao, Zhengmao Hu, Junling Wang, and Zhifan Zhou
- Subjects
Adult ,Male ,Ribosomal Proteins ,0301 basic medicine ,China ,Candidate gene ,Ataxia ,Hereditary spastic paraplegia ,Nonsense mutation ,Pedigree chart ,Consanguinity ,Biology ,Mitochondrial Proteins ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Genetic Testing ,Spinocerebellar Degenerations ,Genetics ,Tripeptidyl-Peptidase 1 ,Spastic Paraplegia, Hereditary ,Homozygote ,DNA Helicases ,Chromosome Mapping ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Disease gene identification ,medicine.disease ,Multifunctional Enzymes ,Pedigree ,030104 developmental biology ,Neurology ,Child, Preschool ,Female ,Neurology (clinical) ,Geriatrics and Gerontology ,medicine.symptom ,RNA Helicases ,030217 neurology & neurosurgery ,ADCK3 - Abstract
Introduction Genetic inheritance plays key roles in patients with ataxia and/or spastic paraplegia in consanguineous families. This study aims to clarify the genetic spectrum of patients with autosomal recessive hereditary ataxia and spastic paraplegias (AR-HA/HSPs) in consanguineous families. Methods A total of 36 AR-HA/HSPs consanguineous pedigrees from China were recruited into this study. Next generation sequencing (NGS), guided by homozygosity mapping (HM), was applied to identify the pathogenic variants in known genes or novel candidate genes. Results We totally made molecular diagnosis in 47.2% (17/36) of AR-HA/HSPs families. Among them, 13 AR-HAs carried pathogenic variants in SETX (n = 4), SACS (n = 2), STUB1, HSD17B4, NEU1, ADCK3, TPP1, PLA2G6 and MTCL1, while four AR-HSPs carried pathogenic variants in SPG11, ZFYVE26, ATP13A2 and ABCD1. One homozygous nonsense mutation in MRPS27 was identified in an AR-HA family, which was potentially a novel candidate gene of AR-HA. Conclusion HM and NGS can serve as an efficient molecular diagnostic tool for AR-HA/HSPs in consanguineous families. Our findings provide a better understanding of genetic architecture of AR-HA/HSPs in consanguinity and broaden the clinical-genetic spectrum of the disease.
- Published
- 2020
- Full Text
- View/download PDF
32. Whole-exome sequencing and homozygosity mapping identify variants in NCOR1 and MAP2K3 associated with non-syndromic congenital heart defects
- Author
-
Mysore R. Savitha, Santhosh Krishnappa, Swati Agarwala, Kavya G, Rahim Gholipoorfeshkecheh, and Nallur B. Ramachandra
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Biology ,Ventricular septal defect ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Gene mapping ,Gene ,Genetics (clinical) ,Exome sequencing ,Genetics ,lcsh:R5-920 ,Mitogen-Activated Protein Kinase 3 ,Haplotype ,Nuclear Corepressor ,In silico validation ,Homozygous regions ,Disease gene identification ,Chromosome 17 (human) ,lcsh:Genetics ,030104 developmental biology ,Whole-exome sequencing ,Haploinsufficiency ,lcsh:Medicine (General) ,030217 neurology & neurosurgery - Abstract
Background Homozygosity mapping is an efficient gene mapping method applicable to recessive disorders. It can detect homozygous segments of identical haplotype structures shared at a higher frequency among ventricular septal defect (VSD) and tetralogy of Fallot (TOF) cases. This study aims to identify the recessive genes involved in congenital heart disease (CHD) cases by homozygosity mapping. A total of 36 CHD cases of Indian origin were recruited based on inclusion and exclusion criteria, disease severity, and hole size. Of these, ten prediagnosed VSD and TOF cases were selected for homozygosity mapping. For in silico validation of variations, overlapping gene variants were analyzed from 26 cases based on pathogenecity and haploinsufficiency scores. Results Genome-wide homozygosity mapping identified 34 homozygous regions with a maximum block length of 80 bp marked for the CHD samples under study. A total of 4863 genes were identified in these 34 homozygous regions, which were present across almost all chromosomes except chromosomes 4, 8, 12, and 13. The homozygosity region found in chromosome 17 revealed genes for CHD manifestation. This homozygous region contained VSD- and TOF-related genes—Nuclear Corepressor 1 (NCOR1) and Mitogen-Activated Protein Kinase Kinase 3 (MAP2K3). In silico validation identified damaging variants for NCOR1 and MAP2K3. Three variants, G207C, C241T, and G244A, were found on exon 2 in the transcript NM_001190438 for NCOR1. Three variants were also found for MAP2K3, namely G194T and C199T in exon 5 and C578T in exon 8 in the transcript NM_002756. All these variants were present in the protein kinase domain. Conclusion Presence of homozygous regions identifies recessive genes leading to disease severity. Defects in recessive genes NCOR1 and MAP2K3 are responsible for abnormal myogenesis, resulting in CHD manifestation.
- Published
- 2020
- Full Text
- View/download PDF
33. Novel nephronophthisis-associated variants reveal functional importance of MAPKBP1 dimerization for centriolar recruitment
- Author
-
Elena Hantmann, Jan Halbritter, Richard Sandford, Melanie Nemitz-Kliemchen, Friedhelm Hildebrandt, Anna Seidel, Ria Schönauer, Nydia Panitz, Daniela A. Braun, Khurrum Shahzad, Matthias Hansen, Wenjun Jin, Anastasia Ertel, Sophie Saunier, Carsten Bergmann, Shirlee Shril, and Alexandre Benmerah
- Subjects
Adult ,0301 basic medicine ,030232 urology & nephrology ,Cell Cycle Proteins ,Nerve Tissue Proteins ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Microtubule ,Nephronophthisis ,medicine ,Humans ,Basal body ,Cilia ,Exome sequencing ,Centrosome ,Polycystic Kidney Diseases ,Cilium ,Intracellular Signaling Peptides and Proteins ,Cell cycle ,medicine.disease ,Disease gene identification ,Fibrosis ,Cell biology ,030104 developmental biology ,Nephrology ,Dimerization - Abstract
Biallelic mutations in MAPKBP1 were recently associated with late-onset cilia-independent nephronophthisis. MAPKBP1 was found at mitotic spindle poles but could not be detected at primary cilia or centrosomes. Here, by identification and characterization of novel MAPKBP1 variants, we aimed at further investigating its role in health and disease. Genetic analysis was done by exome sequencing, homozygosity mapping, and a targeted kidney gene panel while coimmunoprecipitation was used to explore wild-type and mutant protein-protein interactions. Expression of MAPKBP1 in non-ciliated HeLa and ciliated inner medullary collecting duct cells enabled co-localization studies by fluorescence microscopy. By next generation sequencing, we identified two novel homozygous MAPKBP1 splice-site variants in patients with nephronophthisis-related chronic kidney disease. Splice-site analyses revealed truncation of C-terminal coiled-coil domains and patient-derived deletion constructs lost their ability to homodimerize and heterodimerize with paralogous WDR62. While wild-type MAPKBP1 exhibited centrosomal, basal body, and microtubule association, mutant proteins lost the latter and showed reduced recruitment to cell cycle dependent centriolar structures. Wild-type and mutant proteins had no reciprocal influence upon co-expression excluding dominant negative effects. Thus, MAPKBP1 appears to be a novel microtubule-binding protein with cell cycle dependent centriolar localization. Truncation of its coiled-coil domain is enough to abrogate its dimerization and results in severely disturbed intracellular localizations. Delineating the impact of impaired dimerization on cell cycle regulation and intracellular kidney signaling may provide new insights into common mechanisms of kidney degeneration. Thus, due to milder clinical presentation, MAPKBP1-associated nephronophthisis should be considered in adult patients with otherwise unexplained chronic kidney disease.
- Published
- 2020
- Full Text
- View/download PDF
34. Homozygosity mapping and whole exome sequencing provide exact diagnosis of Cohen syndrome in a Saudi family
- Author
-
Ahmed Almatrafi, Sibtain Afzal, Peter Nürnberg, Jamil A. Hashmi, Khushnooda Ramzan, Fatima Fadhli, Holger Thiele, and Sulman Basit
- Subjects
Male ,Genotype ,Developmental Disabilities ,Saudi Arabia ,Vesicular Transport Proteins ,Biology ,Polymorphism, Single Nucleotide ,Frameshift mutation ,Fingers ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Intellectual Disability ,Exome Sequencing ,Myopia ,medicine ,Humans ,Obesity ,Global developmental delay ,Frameshift Mutation ,Indel ,Exome sequencing ,Genetics ,Cohen syndrome ,Homozygote ,Retinal Degeneration ,Chromosome Mapping ,General Medicine ,Disease gene identification ,medicine.disease ,Pedigree ,VPS13B ,Pediatrics, Perinatology and Child Health ,Microcephaly ,Muscle Hypotonia ,Female ,Neurology (clinical) ,Vesicle-mediated transport ,030217 neurology & neurosurgery - Abstract
Background Cohen syndrome (CS) is a rare multi-system autosomal recessive disorder with a high prevalence in the Finnish population. Clinical features of Finnish-type CS are homogeneous, however, in non-Finnish populations, CS diagnosis is challenging due to broad phenotypic variability. Methods We studied a consanguineous family having three affected individuals with clinical features of severe intellectual disability and global developmental delay. Clinical diagnosis of the phenotype could not be established based on the features. Therefore, whole genome SNP genotyping and whole exome sequencing (WES) were performed on DNA samples from affected and unaffected family members. Results Homozygosity mapping identified a shared loss of heterozygosity region on chromosome 8q22.1-q22.3 and WES data analysis revealed an insertion-deletion (indel) mutation (c.11519_11521delCAAinsT) in the VPS13B gene. The indel is predicted to cause a frameshift resulting in a premature termination of the VPS13B protein (NP_060360.3:p.Pro3840Leufs*2). Conclusion VPS13B encodes a giant transmembrane protein called vacuolar protein sorting 13 homolog B. VPS13B is known to play a role in the glycosylation of Golgi proteins and in endosomal-lysosomal trafficking. Moreover, it is thought to function in vesicle mediated transport and sorting of proteins within the cell. The mechanism by which abnormalities of the VPS13B protein lead to the phenotype of CS is currently unknown. Here, in this study, we successfully established a clinical diagnosis of CS cases from a family using genomic analyses. Clinical re-examination of the patients revealed characteristic ocular abnormalities.
- Published
- 2020
- Full Text
- View/download PDF
35. Advancing Charcot-Marie-Tooth disease diagnostics, through the UK 100,000 Genomes Project
- Author
-
Henry Houlden, Menelaos Pipis, and Mary M. Reilly
- Subjects
Whole genome sequencing ,medicine.medical_specialty ,Public health ,Genomics ,Disease ,Disease gene identification ,Data science ,Genome ,Structural variation ,Geography ,Biorepository ,Genetics ,medicine ,Genetics (clinical) - Abstract
Whole genome sequencing (WGS) is regarded by many as the pinnacle of contemporary molecular genetic testing, and has only been possible because of the rapid development and roll-out of next-generation sequencing technologies. It provides a phenotype-agnostic analysis of the genome and has important advantages compared to other techniques including a consistent coverage across the coding and non-coding genome, the application of high resolution homozygosity mapping and the ability to detect and highlight structural variation. Realising this potential and with a bid to sequence 100,000 genomes, the UK rolled out the 100,000 Genomes Project as a proof of concept of integrating genomics in the national health service. Participants with cancer and rare diseases enrolled in the project whose infrastructure comprises of a central national biorepository and 13 regional genomic medicine centres where clinicians, geneticists and other scientists work as part of a multidisciplinary team. Amongst participants are also patients with genetically unclassified Charcot-Marie-Tooth disease who have benefited substantially from improved diagnostic rates and many more stand to benefit as the analysis of genomic data is ongoing. WGS is an important tool as we head towards more personalised medicine and in our quest to improve public health and treat and where possible prevent disease.
- Published
- 2020
- Full Text
- View/download PDF
36. Usher syndrome and Nebulin‐associated myopathy in a single patient due to variants inMYO7AandNEB
- Author
-
Arjan P.M. de Brouwer, Ana Maria Fortuna, Paula Jorge, Ana L. Gonçalves, Isabel Marques, Rosário Santos, Manuel Melo Pires, Ana Rita Soares, and Nuno Maia
- Subjects
Medicine (General) ,MYO7A ,Usher syndrome ,Case Report ,Case Reports ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Nebulin ,R5-920 ,0302 clinical medicine ,otorhinolaryngologic diseases ,Medicine ,Myopathy ,Gene ,Exome sequencing ,Genetics ,biology ,business.industry ,NEB ,General Medicine ,medicine.disease ,Disease gene identification ,Phenotype ,eye diseases ,Nebulin‐associated myopathy ,homozygosity mapping ,030220 oncology & carcinogenesis ,biology.protein ,medicine.symptom ,business - Abstract
In a patient with Usher syndrome and atypical muscle complaints, we have identified two separate variants in MYO7A andNEB genes by exome sequencing. The homozygous variants in these two recessive genes could explain the full phenotype of our patient., In a patient with Usher syndrome and atypical muscle complaints, we have identified two separate variants in MYO7A and NEB genes by exome sequencing. The homozygous variants in these two recessive genes could explain the full phenotype of our patient.
- Published
- 2020
- Full Text
- View/download PDF
37. A case of Usher syndrome type IIA caused by a rare USH2A homozygous frameshift variant with maternal uniparental disomy (UPD) in a Chinese family
- Author
-
Hongbin Lv, Shiyi Shen, Jiewen Fu, Junjiang Fu, and Jingliang Cheng
- Subjects
Male ,0301 basic medicine ,Proband ,frameshift mutation ,Usher syndrome ,DNA Mutational Analysis ,0302 clinical medicine ,USH2A gene ,Exome sequencing ,Genetics ,Sanger sequencing ,Extracellular Matrix Proteins ,Homozygote ,uniparental disomy (UPD) ,Disease gene identification ,Pedigree ,whole exome sequencing (WES) ,Phenotype ,homozygosity mapping ,Child, Preschool ,030220 oncology & carcinogenesis ,symbols ,Molecular Medicine ,Original Article ,Female ,Maternal Inheritance ,Usher Syndromes ,Adult ,China ,congenital, hereditary, and neonatal diseases and abnormalities ,short tandem repeat ,Genetic counseling ,Biology ,Frameshift mutation ,03 medical and health sciences ,symbols.namesake ,Asian People ,Retinitis pigmentosa ,otorhinolaryngologic diseases ,medicine ,Humans ,Genetic Association Studies ,Whole Genome Sequencing ,Computational Biology ,Original Articles ,Cell Biology ,Uniparental Disomy ,medicine.disease ,eye diseases ,030104 developmental biology ,usher syndrome type IIA - Abstract
Usher syndrome encompasses a group of genetically and clinically heterogeneous autosomal recessive disorders with hearing deficiencies and retinitis pigmentosa. The mechanisms underlying the Usher syndrome are highly variable. In the present study, a Chinese family with Usher syndrome was recruited. Whole exome sequencing (WES), Sanger sequencing, homozygosity mapping, short tandem repeat (STR) analysis and segregation analysis were performed. Functional domains of the pathogenic variant for USH2A were analysed. We identified a homozygous frameshift variant c.99_100insT (p.Arg34Serfs*41) in the USH2A gene in the proband that showed discordant segregation in the father. Further homozygosity mapping and STR analysis identified an unusual homozygous variant of proband that originated from maternal uniparental disomy (UPD). The p.Arg34Serfs*41 variant produced a predicted truncated protein that removes all functional domains of USH2A. The variant was not included in the 1000 Human Genomes Project database, ExAC database, HGMD or gnomAD database, but was included in the ClinVar databases as pathogenic. Although USH2A is an autosomal recessive disease, the effects of UPD should be informed in genetic counselling since the recurrence risk of an affected child is greatly reduced when the disease is due to the UPD mechanism. To test potential patients, WES, combined with STR analysis and homozygosity mapping, provides an accurate and useful strategy for genetic diagnosis. In summary, our discoveries can help further the understanding of the molecular pathogenesis of Usher syndrome type IIA to advance the prevention, diagnosis and therapy for this disorder.
- Published
- 2020
- Full Text
- View/download PDF
38. A Novel SEMA3G Mutation in Two Siblings Affected by Syndromic GnRH Deficiency
- Author
-
Maria Vittoria Corridori, Roberto Oleari, Valentina Andre, Ludovica Cotellessa, Carles Gaston-Massuet, Anna Cariboni, Hellmut G. Augustin, Khalid Hussain, Lise Roth, Sophia Tahir, Valeria Scagliotti, Lisa Benedetta De Martini, Ivano Eberini, Simona Gulli, Antonella Lettieri, Chiara Parravicini, Francesco Bedogni, and Alyssa Paganoni
- Subjects
Male ,Hypothalamo-Hypophyseal System ,medicine.medical_specialty ,Developmental Disabilities ,Endocrinology, Diabetes and Metabolism ,In silico ,ved/biology.organism_classification_rank.species ,030209 endocrinology & metabolism ,Semaphorins ,Biology ,030218 nuclear medicine & medical imaging ,Craniofacial Abnormalities ,Gonadotropin-Releasing Hormone ,Consanguinity ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Endocrinology ,Semaphorin ,Hypogonadotropic hypogonadism ,Intellectual Disability ,Internal medicine ,medicine ,Animals ,Humans ,Model organism ,Cells, Cultured ,Exome sequencing ,Genetics ,GnRH Neuron ,Endocrine and Autonomic Systems ,ved/biology ,Hypogonadism ,Siblings ,Homozygote ,Genetic disorder ,Syndrome ,Disease gene identification ,medicine.disease ,Pedigree - Abstract
Introduction: Gonadotropin-releasing hormone (GnRH) deficiency causes hypogonadotropic hypogonadism (HH), a rare genetic disorder that impairs sexual reproduction. HH can be due to defective GnRH-secreting neuron development or function and may be associated with other clinical signs in overlapping genetic syndromes. With most of the cases being idiopathic, genetics underlying HH is still largely unknown. Objective: To assess the contribution of mutated Semaphorin 3G (SEMA3G) in the onset of a syndromic form of HH, characterized by intellectual disability and facial dysmorphic features. Method: By combining homozygosity mapping with exome sequencing, we identified a novel variant in the SEMA3G gene. We then applied mouse as a model organism to examine SEMA3Gexpression and its functional requirement in vivo. Further, we applied homology modelling in silico and cell culture assays in vitro to validate the pathogenicity of the identified gene variant. Results: We found that (i) SEMA3G is expressed along the migratory route of GnRH neurons and in the developing pituitary, (ii) SEMA3G affects GnRH neuron development, but is redundant in the adult hypothalamic-pituitary-gonadal axis, and (iii) mutated SEMA3G alters binding properties in silico and in vitro to its PlexinA receptors and attenuates its effect on the migration of immortalized GnRH neurons. Conclusion: In silico, in vitro, and in vivo models revealed that SEMA3G regulates GnRH neuron migration and that its mutation affecting receptor selectivity may be responsible for the HH-related defects.
- Published
- 2020
- Full Text
- View/download PDF
39. Mutation in ALOX12B likely cause of POI and also ichthyosis in a large Iranian pedigree
- Author
-
Mohammad Masoud Rahimi Bidgoli, Judith Fischer, Faezeh Darki, Mostafa Mirshams Shahshahani, Elahe Elahi, Ashraf Moini, Afagh Alavi, and Davood Zare-Abdollahi
- Subjects
Adult ,0106 biological sciences ,0301 basic medicine ,Genetic Linkage ,Anion Transport Proteins ,Menopause, Premature ,ALOX12B ,Locus (genetics) ,Iran ,Primary Ovarian Insufficiency ,Biology ,Arachidonate 12-Lipoxygenase ,Premature ovarian insufficiency ,01 natural sciences ,Consanguinity ,03 medical and health sciences ,Genetic linkage ,Exome Sequencing ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Molecular Biology ,Exome sequencing ,Ichthyosis ,Homozygote ,General Medicine ,Lipid Metabolism ,Disease gene identification ,medicine.disease ,Human genetics ,Pedigree ,030104 developmental biology ,Mutation ,Female ,010606 plant biology & botany - Abstract
Premature ovarian insufficiency (POI) is a clinically and etiologically heterogeneous disorder characterized by menstrual irregularities and elevated levels of FSH before age of 40 years. Genetic anomalies are among the recognized causes of POI. Here, we aimed to identify the genetic cause of POI in an inbred pedigree with nine POI and two ichthyosis-affected members. Inheritance of POI and ichthyosis were, respectively, dominant and recessive. Reproduction-related information and measurements of relevant hormones were obtained. Genetic studies included homozygosity mapping, linkage analysis, exome sequencing, and screening of candidate variants. A mutation within ALOX12B, which is a known ichthyosis causing gene, was identified as cause of ichthyosis. ALOX12B encodes a protein involved in steroidogenesis and lipid metabolism. Considering the importance of steroidogenesis in reproduction functions, the possibility that the ALOX12B mutation is also cause of POI was considered. Screenings showed that the mutation segregated with POI status. Linkage analysis with respect to POI identified a single strongly linked locus (LOD > 3) that includes ALOX12B. Exome sequencing on POI-affected females identified the mutation in ALOX12B and also a sequence variation in SPNS2 within the linked locus. A possible contribution of the SPNS2 variation to POI was not strictly ruled out, but various data presented in the text including reported association of variations in related gene ALOX12 with menopause-age and role of ALOX12B in atretic bovine follicle formation argue in favor of ALOX12B. It is, therefore, concluded that the mutation in ALOX12B is the likely cause of POI in the pedigree.
- Published
- 2020
- Full Text
- View/download PDF
40. A novel pathogenic mutation on Interleukin-7 receptor leading to severe combined immunodeficiency identified with newborn screening and whole exome sequencing
- Author
-
Chao Neng Cheng, Cheng Yu Liao, Hui Wen Yu, Ching Wei Lin, Jiann Shiuh Chen, Peng Chieh Chen, and Chi Chang Shieh
- Subjects
0301 basic medicine ,Microbiology (medical) ,Proband ,030106 microbiology ,lcsh:QR1-502 ,Human leukocyte antigen ,Consanguinity ,lcsh:Microbiology ,Frameshift mutation ,Interleukin-7 Receptor alpha Subunit ,03 medical and health sciences ,Neonatal Screening ,0302 clinical medicine ,Exome Sequencing ,medicine ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,Frameshift Mutation ,Exome sequencing ,Severe combined immunodeficiency ,Newborn screening ,General Immunology and Microbiology ,business.industry ,Homozygote ,Infant, Newborn ,Sequence Analysis, DNA ,General Medicine ,medicine.disease ,Disease gene identification ,Pedigree ,Infectious Diseases ,Immunology ,Female ,Severe Combined Immunodeficiency ,business - Abstract
Background: Patients with severe combined immunodeficiency (SCID), which is caused by genetic defects in immune-related genes involved in the development or activation of the adaptive immune system, often died in infancy due to severe infections before definite molecular diagnosis could be made. Although recent improvement in early diagnosis has been achieved by newborn screening, the genetic basis of many of the patients is still unknown. Methods: Here we performed whole exome sequencing (WES) to investigate the underlying genetic causes of SCID in a proband identified with newborn screening. Inheritance of the mutation was confirmed with targeted sequencing of the parents. Homozygosity mapping from the WES was used to investigate the consanguinity of the parents. Immunoblotting was used to confirm the loss of expression of the mutant protein. Results: A novel homozygous frameshift mutation of IL7R was identified through WES. Both parents are carriers for this 1-bp deletion. HLA typing and exome-wide homozygous stretch mapping suggested that the parents are consanguineous. Immunoblotting showed no expression of IL7Rα isoform in the whole blood sample of the proband. The proband received peripheral blood stem cell transplantation and her general condition became stable. Our results suggest that IL7R is essential for T cell development but dispensable for the development of certain human NK cells B cells and suggest that WES can be a useful tool for precise genetic diagnosis of SCID following newborn screening in the index patient without the need to screen other members of the whole family. Keywords: Severe combined immunodeficiency, Exome sequencing, IL7R
- Published
- 2020
41. Next‐generation sequencing and the evolution of data sharing
- Author
-
Nara Sobreira and Ada Hamosh
- Subjects
0301 basic medicine ,ved/biology.organism_classification_rank.species ,Computational biology ,030105 genetics & heredity ,Biology ,DNA sequencing ,Evolution, Molecular ,03 medical and health sciences ,Databases, Genetic ,Genetics ,Humans ,Genetic Predisposition to Disease ,Model organism ,Gene ,Genetics (clinical) ,Exome sequencing ,Disease gene ,Information Dissemination ,ved/biology ,Genetic Diseases, Inborn ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Disease gene identification ,Phenotype ,Data sharing ,030104 developmental biology - Abstract
Disease gene identification often relies on identifying multiple affected individuals with similar phenotypes and candidate variants in the same gene. Phenotypic and genomic data sharing tools have facilitated connections that led to novel disease gene discoveries and better characterization and recognition of rare diseases. Additionally, data sharing has evolved. From gene-based matches to variant-level information with increasing use of phenotypic information. We expect that these initiatives will continue to expand in the future affording clinicians, researchers, and most importantly, patients and their families faster and more comprehensive answers.
- Published
- 2021
- Full Text
- View/download PDF
42. Phenotypic spectrum and genomics of undiagnosed arthrogryposis multiplex congenital
- Author
-
Ivo Gut, Sarah Grotto, Céline Bellesme, Arnold Munnich, Cyril Gitiaux, Jeanne Amiel, Chloé Quélin, Annie Laquerrière, Suonavy Khung, Hanitra Ranjatoelina-Randrianaivo, Luc Rigonnot, Christine Francannet, Loic Quevarec, Jérôme Bouligand, Fabienne Prieur, Alexandra Benachi, Valérie Cormier-Daire, Laurence Perrin, Judith Melki, Pierre-Simon Jouk, Flora Nolent, Tania Attié-Bitach, Delphine Héron, Marie-Line Jacquemont, Claire Beneteau, Fabien Guimiot, Laetitia Lambert, Sandra Mercier, Valérie Biancalana, Fanny Laffargue, Elise Boucher, Jean-Louis Bessereau, P. Landrieu, Annick Toutain, Alain Verloes, Alice Goldenberg, Philippe Latour, Dominique Martin-Coignard, Anne Guiochon-Mantel, Dan Mejlachowicz, Damien Sternberg, Haluk Topaloglu, Bruno Eymard, Géraldine Viot, Catherine Fallet-Bianco, Julien Saada, Isabelle Desguerre, Marie-Hélène Saint-Frison, Catherine Vincent-Delorme, Sophie Blesson, Radka Stoeva, Alexandre J. Vivanti, Martine Bucourt, Pascaline Letard, Jérome Maluenda, Laurence Loeuillet, Lionel Van Maldergem, Didier Lacombe, Marcel Tawk, Michèle Granier, Stanislas Lyonnet, Anne-Lise Delezoide, Andrée Delahaye-Duriez, André Mégarbané, Marie Gonzales, Florence Petit, Juliette Piard, Laurence Faivre, Helene Verhelst, Bettina Bessières, Sabine Sigaudy, Sandra Whalen, Valérie Layet, Yline Capri, Fanny Pelluard, Emanuela Abiusi, Klaus Dieterich, Marie Vincent, Marine Legendre, Dana Jaber, Romulus Grigorescu, Florent Marguet, Eric Bieth, Helge Amthor, Christine Barnerias, Estelle Colin, Laetitia Trestard, Mathilde Nizon, Jelena Martinovic, Daniel Amram, and Nicoletta Resta
- Subjects
musculoskeletal diseases ,Artrogriposi múltiple congènita ,Settore BIO/18 - GENETICA ,human genetics ,neuromuscular diseases ,Genomics ,Biology ,CONTRACTURES ,CLASSIFICATION ,diseases ,symbols.namesake ,Diagnòstic ,Gene mapping ,arthrogryposis multiplex congenita ,Exome Sequencing ,OF-FUNCTION MUTATIONS ,Genetics ,Medicine and Health Sciences ,genomics ,Humans ,Genetics (clinical) ,Exome sequencing ,Arthrogryposis ,Sanger sequencing ,Arthrogryposis multiplex congenita ,Genetic heterogeneity ,SPINAL MUSCULAR-ATROPHY ,Proteins ,nervous system malformations ,DYSTROPHY ,Disease gene identification ,GENE ,Human genetics ,Pedigree ,ETIOLOGY ,Phenotype ,symbols ,neuromuscular ,Genètica ,Transcription Factors - Abstract
BackgroundArthrogryposis multiplex congenita (AMC) is characterised by congenital joint contractures in two or more body areas. AMC exhibits wide phenotypic and genetic heterogeneity. Our goals were to improve the genetic diagnosis rates of AMC, to evaluate the added value of whole exome sequencing (WES) compared with targeted exome sequencing (TES) and to identify new genes in 315 unrelated undiagnosed AMC families.MethodsSeveral genomic approaches were used including genetic mapping of disease loci in multiplex or consanguineous families, TES then WES. Sanger sequencing was performed to identify or validate variants.ResultsWe achieved disease gene identification in 52.7% of AMC index patients including nine recently identified genes (CNTNAP1, MAGEL2, ADGRG6, ADCY6, GLDN, LGI4, LMOD3, UNC50 and SCN1A). Moreover, we identified pathogenic variants in ASXL3 and STAC3 expanding the phenotypes associated with these genes. The most frequent cause of AMC was a primary involvement of skeletal muscle (40%) followed by brain (22%). The most frequent mode of inheritance is autosomal recessive (66.3% of patients). In sporadic patients born to non-consanguineous parents (n=60), de novo dominant autosomal or X linked variants were observed in 30 of them (50%).ConclusionNew genes recently identified in AMC represent 21% of causing genes in our cohort. A high proportion of de novo variants were observed indicating that this mechanism plays a prominent part in this developmental disease. Our data showed the added value of WES when compared with TES due to the larger clinical spectrum of some disease genes than initially described and the identification of novel genes.
- Published
- 2022
43. Progressive Myoclonus Epilepsies
- Author
-
Edoardo Ferlazzo, Laura Licchetta, Alessandro Filla, Samuel F. Berkovic, Roberto Michelucci, Paolo Tinuper, Martina Fanella, Silvana Franceschetti, Anna Teresa Giallonardo, Carolina Courage, Federico Zara, Antonio Gambardella, Teresa Anna Cantisani, Patrizia Riguzzi, Barbara Castellotti, Cinzia Gellera, Pasquale Striano, Carlo Di Bonaventura, Tiziana Granata, Melanie Bahlo, Laura Canafoglia, Karen Oliver, Angela La Neve, Anna-Elina Lehesjoki, Adriana Magaudda, HUSLAB, Medicum, Department of Medical and Clinical Genetics, Canafoglia L., Franceschetti S., Gambardella A., Striano P., Giallonardo A.T., Tinuper P., Di Bonaventura C., Michelucci R., Ferlazzo E., Granata T., Magaudda A., Licchetta L., Filla A., la Neve A., Riguzzi P., Cantisani T.A., Fanella M., Castellotti B., Gellera C., Bahlo M., Zara F., Courage C., Lehesjoki A.-E., Oliver K.L., and Berkovic S.F.
- Subjects
Biology ,DNA sequencing ,3124 Neurology and psychiatry ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Sibling ,Gene ,myoclonus epilepsy ,Genetics (clinical) ,030304 developmental biology ,Genetic testing ,Genetics ,0303 health sciences ,medicine.diagnostic_test ,3112 Neurosciences ,next generation sequency ,Disease gene identification ,3. Good health ,CHD2 ,3121 General medicine, internal medicine and other clinical medicine ,Epilepsy syndromes ,Neurology (clinical) ,medicine.symptom ,Myoclonus ,030217 neurology & neurosurgery - Abstract
Background and ObjectivesTo assess the current diagnostic yield of genetic testing for the progressive myoclonus epilepsies (PMEs) of an Italian series described in 2014 where Unverricht-Lundborg and Lafora diseases accounted for ∼50% of the cohort.MethodsOf 47/165 unrelated patients with PME of indeterminate genetic origin, 38 underwent new molecular evaluations. Various next-generation sequencing (NGS) techniques were applied including gene panel analysis (n = 7) and/or whole-exome sequencing (WES) (WES singleton n = 29, WES trio n = 7, and WES sibling n = 4). In 1 family, homozygosity mapping was followed by targeted NGS. Clinically, the patients were grouped in 4 phenotypic categories: “Unverricht-Lundborg disease-like PME,” “late-onset PME,” “PME plus developmental delay,” and “PME plus dementia.”ResultsSixteen of 38 (42%) unrelated patients reached a positive diagnosis, increasing the overall proportion of solved families in the total series from 72% to 82%. Likely pathogenic variants were identified in NEU1 (2 families), CERS1 (1 family), and in 13 nonfamilial patients in KCNC1 (3), DHDDS (3), SACS, CACNA2D2, STUB1, AFG3L2, CLN6, NAXE, and CHD2. Across the different phenotypic categories, the diagnostic rate was similar, and the same gene could be found in different phenotypic categories.DiscussionThe application of NGS technology to unsolved patients with PME has revealed a collection of very rare genetic causes. Pathogenic variants were detected in both established PME genes and in genes not previously associated with PME, but with progressive ataxia or with developmental encephalopathies. With a diagnostic yield >80%, PME is one of the best genetically defined epilepsy syndromes.
- Published
- 2021
44. A knockout mutation associated with juvenile paroxysmal dyskinesia in Markiesje dogs indicates SOD1 pleiotropy
- Author
-
Mandigers, P J J, Van Steenbeek, F G, Bergmann, W, Vos-Loohuis, M, Leegwater, P A, CS_Genetics, Interne geneeskunde GD, Dep Clinical Sciences, dCSCA AVR, Dep Pathobiologie, VPDC pathologie, dPB CR, dCSCA RMSC-1, CS_Genetics, Interne geneeskunde GD, Dep Clinical Sciences, dCSCA AVR, Dep Pathobiologie, VPDC pathologie, dPB CR, and dCSCA RMSC-1
- Subjects
Male ,040301 veterinary sciences ,SOD1 ,Physiology ,Genes, Recessive ,Biology ,Frameshift mutation ,0403 veterinary science ,03 medical and health sciences ,Dogs ,Superoxide Dismutase-1 ,0302 clinical medicine ,Chorea ,medicine ,Genetics ,Animals ,Genetics(clinical) ,Dog Diseases ,Amyotrophic lateral sclerosis ,Frameshift Mutation ,Genetics (clinical) ,Original Investigation ,Homozygote ,Chromosome Mapping ,High-Throughput Nucleotide Sequencing ,Genetic Pleiotropy ,04 agricultural and veterinary sciences ,Paroxysmal dyskinesia ,Disease gene identification ,medicine.disease ,Penetrance ,Pedigree ,nervous system diseases ,Mutation (genetic algorithm) ,Female ,Age of onset ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
A juvenile form of paroxysmal dyskinesia segregated in the Markiesje dog breed. Affected pups exhibited clinical signs of a severe tetraparesis, dystonia, cramping and falling over when trying to walk. In most cases, the presentation deteriorated within weeks and elective euthanasia was performed. Pedigree analysis indicated autosomal recessive inheritance. Genome-wide association and homozygosity mapping of 5 affected dogs from 3 litters identified the associated locus on chromosome 31 in the region of SOD1. The DNA sequence analysis of SOD1 showed that the patients were homozygous for a frameshift mutation in the fourth codon. None of the other analyzed dogs of the breed was homozygous for the mutation, indicating full penetrance of the genetic defect. Mutations in SOD1 are known to cause recessive degenerative myelopathy in middle-aged dogs with low penetrance and dominant amyotrophic lateral sclerosis in humans with variable age of onset. Our findings are similar to recent observations in human patients that a loss of function mutation in SOD1 leads to a juvenile neurologic disease distinct from amyotrophic lateral sclerosis.
- Published
- 2021
45. Perceptron ensemble of graph-based positive-unlabeled learning for disease gene identification.
- Author
-
Jowkar, Gholam-Hossein and Mansoori, Eghbal G.
- Subjects
- *
MEDICAL genetics , *PERCEPTRONS , *GRAPH theory , *BIOINFORMATICS , *BIOMEDICAL engineering , *PHENOTYPES - Abstract
Identification of disease genes, using computational methods, is an important issue in biomedical and bioinformatics research. According to observations that diseases with the same or similar phenotype have the same biological characteristics, researchers have tried to identify genes by using machine learning tools. In recent attempts, some semi-supervised learning methods, called positive-unlabeled learning, is used for disease gene identification. In this paper, we present a Perceptron ensemble of graph-based positive-unlabeled learning (PEGPUL) on three types of biological attributes: gene ontologies, protein domains and protein-protein interaction networks. In our method, a reliable set of positive and negative genes are extracted using co-training schema. Then, the similarity graph of genes is built using metric learning by concentrating on multi-rank-walk method to perform inference from labeled genes. At last, a Perceptron ensemble is learned from three weighted classifiers: multilevel support vector machine, k -nearest neighbor and decision tree. The main contributions of this paper are: (i) incorporating the statistical properties of gene data through choosing proper metrics, (ii) statistical evaluation of biological features, and (iii) noise robustness characteristic of PEGPUL via using multilevel schema. In order to assess PEGPUL, we have applied it on 12950 disease genes with 949 positive genes from six class of diseases and 12001 unlabeled genes. Compared with some popular disease gene identification methods, the experimental results show that PEGPUL has reasonable performance. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
46. Homozygous MEFV Gene Variant and Pyrin-Associated Autoinflammation With Neutrophilic Dermatosis: A Family With a Novel Autosomal Recessive Mode of Inheritance
- Author
-
Mehdi Yousefi, Vahid Ziaee, Mohammad-Sadegh Soltani Zangbar, Leila Youssefian, Sirous Zeinali, Soroush Shahrokh, Amir Hossein Saeidian, Kambiz Kamyab Hesari, Jouni Uitto, Hamidreza Mahmoudi, Hassan Vahidnezhad, Nima Parvaneh, and Bahar Ashjaei
- Subjects
Proband ,Male ,Familial Mediterranean fever ,Dermatitis ,Dermatology ,Iran ,Pyrin domain ,medicine ,Missense mutation ,Humans ,Child ,Exome sequencing ,business.industry ,Brief Report ,Homozygote ,Pyrin ,Disease gene identification ,medicine.disease ,MEFV ,Familial Mediterranean Fever ,Child, Preschool ,Immunology ,Mutation ,Prednisolone ,Female ,business ,medicine.drug - Abstract
IMPORTANCE: Pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND) is a monogenic autoinflammatory disorder with autosomal dominant inheritance and has been associated with monoallelic p.Ser242Arg and p.Glu244Lys variations in the MEFV gene. This dermatosis shares clinical features and pathogenesis with familial Mediterranean fever, although it is a clinically distinct entity. OBJECTIVE: To identify the genetic basis of PAAND in a consanguineous family with 2 affected children and to prescribe an effective genotype-guided treatment. DESIGN, SETTING, AND PARTICIPANTS: This case series study examined 2 siblings who presented with clinical features of PAAND. We sought the genetic basis of this disease with trio whole exome sequencing (trio-WES). Genome-wide homozygosity mapping provided additional evidence for causality of a sequence variant identified by trio-WES. MAIN OUTCOMES AND MEASURES: Association of a biallelic MEFV variation with a new form of autosomal recessive PAAND was documented by genetic analysis. Response to treatment with colchicine and a low-dose steroid was assessed clinically and experimentally. RESULTS: Two siblings, a girl (proband; age 5 years) and a boy (age 2.5 years) of Iranian-Azeri ancestry born to first-cousin consanguineous parents presented with clinical features of PAAND—recurrent episodes of maculopapular and pustular rash, gastrointestinal involvement resembling inflammatory bowel disease, and intussusception with generalized mesenteric lymphadenitis. A trio-WES test detected a previously unreported homozygous missense variation, p.Ser242Gly, in both patients’ MEFV gene. Genome-wide homozygosity mapping revealed shared regions of homozygosity in the patients’ DNA, including 1 on chromosome 16 harboring MEFV. Whole transcriptome sequencing by RNA-sequencing revealed that the variant MEFV transcript, among the inflammasome-associated transcripts, was most upregulated, and the cell-cell receptor interaction and innate immune system pathways were most positively enriched. Under the guidance of MEFV genotype, treatment with colchicine (1 mg/d) and low-dose prednisolone (2.5 mg every other day) was started, and the patients responded well. CONCLUSIONS AND RELEVANCE: This case series study demonstrated successful genotype-guided treatment with colchicine and low-dose prednisolone, a low-cost therapeutic option with minimal adverse effects, in patients with a novel form of autosomal recessive PAAND. This case report examines the genetic basis of PAAND in a consanguineous family with 2 affected children and seeks to prescribe an effective genotype-guided treatment.
- Published
- 2021
47. A recurrent rare intronic variant in CAPN3 alters mRNA splicing and causes autosomal recessive limb-girdle muscular dystrophy-1 in three Pakistani pedigrees
- Author
-
Muhammad Ikram Ullah, Tahir N. Khan, Maimoona Siddiqui, Gaurav V. Harlalka, Erica E. Davis, Talia Akram, Zaineb Akram, Chunyu Liu, Muhammad Jameel, Arsalan Ahmad, Kamal Khan, Sarmad Mehmood, Barry A. Chioza, Muhammad Jawad Hassan, Humayoon Shafique Satti, Raees Khan, Emma A. Baple, Andrew H. Crosby, Shahid Mahmood Baig, Belqees Yawar Faiz, and Feng Zhang
- Subjects
Genetics ,Weakness ,Calpain ,Haplotype ,Muscle Proteins ,Pedigree chart ,Biology ,Disease gene identification ,medicine.disease ,Exon ,Muscular Dystrophies, Limb-Girdle ,RNA splicing ,Mutation ,medicine ,Humans ,Pakistan ,RNA, Messenger ,medicine.symptom ,Genetics (clinical) ,Founder effect ,Limb-girdle muscular dystrophy - Abstract
Autosomal recessive limb-girdle muscular dystrophy-1 (LGMDR1) is an autosomal recessive disorder characterized by progressive weakness of the proximal limb and girdle muscles. Biallelic mutations in CAPN3 are reported frequently to cause LGMDR1. Here, we describe 11 individuals from three unrelated consanguineous families that present with typical features of LGMDR1 that include proximal muscle wasting, weakness of the upper and lower limbs, and elevated serum creatine kinase. Whole-exome sequencing identified a rare homozygous CAPN3 variant near the exon 2 splice donor site that segregates with disease in all three families. mRNA splicing studies showed partial retention of intronic sequence and subsequent introduction of a premature stop codon (NM_000070.3: c.379 + 3A>G; p.Asp128Glyfs*15). Furthermore, we observe reduced CAPN3 expression in primary dermal fibroblasts derived from an affected individual, suggesting instability and/or nonsense-mediated decay of mutation-bearing mRNA. Genome-wide homozygosity mapping and single-nucleotide polymorphism analysis identified a shared haplotype and supports a possible founder effect for the CAPN3 variant. Together, our data extend the mutational spectrum of LGMDR1 and have implications for improved diagnostics for individuals of Pakistani origin.
- Published
- 2021
48. Identification of disease genes and assessment of eye-related diseases caused by disease genes using JMFC and GDLNN
- Author
-
S. R. Nirmala and Samar Jyoti Saikia
- Subjects
Eye Diseases ,Biomedical Engineering ,Bioengineering ,Feature selection ,General Medicine ,Computational biology ,Disease ,Biology ,Disease gene identification ,Computer Science Applications ,Human-Computer Interaction ,Mutation (genetic algorithm) ,Cluster Analysis ,Humans ,Human genome ,Identification (biology) ,sense organs ,Neural Networks, Computer ,Gene ,Function (biology) ,Algorithms - Abstract
Early detection of disease genes helps humans to recover from certain gene-related diseases, like genetic eye diseases. This work identifies the possibility of eye diseasesfor the disease genes utilizing a Gaussian-activation function (G)-centric deeplearning neural network (GDLNN) model. In this work, human genes are selected by computing structural similarity and genes are clustered as disease genesand normal genes by using the JMFC clustering algorithm. Levy flight and Crossover and Mutation (LCM) centric Chicken Swarm Optimization (LCM-CSO) is employed for feature selection and GDLNN classifies the eye-related diseases for the input genes using the selected features.
- Published
- 2021
49. Homozygosity mapping coupled with whole-exome sequencing and protein modelling identified a novel missense mutation in GUCY2D in a consanguineous Pakistani family with Leber congenital amaurosis
- Author
-
John B. Vincent, Saadullah Khan, Muhammad Zubair, Ricardo Harripaul, Muhammad Muzammal, Muhammad Faheem, Muzammil Ahmad Khan, Abdul Haleem Shah, Sumra Wajid Abbasi, and Hadia Gul
- Subjects
Genetics ,Sanger sequencing ,genetic structures ,Genetic heterogeneity ,Biology ,Disease gene identification ,eye diseases ,SNP genotyping ,symbols.namesake ,Mutation (genetic algorithm) ,symbols ,Missense mutation ,GUCY2D ,Exome sequencing - Abstract
Leber congenital amaurosis (LCA) is a rare form of early onset vision loss or blindness due to retinal dystrophy. This condition is characterized by early vision loss, nystagmus and severe retinal dysfunction. To date, genetic studies have reported 19 genes to be associated with autosomal recessive LCA, most of which are involved in the retinal morphology and the physiology of the phototransduction pathway. In the current study, a large consanguineous family segregating congenital blindness was ascertained from the Dera Ismail Khan region of Pakistan. Genetic analysis was performed through genomewide SNP genotyping (for homozygosity-by-descent mapping), whole-exome sequencing (for mutation identification) and Sanger sequencing (for segregation analysis). In silico structural predictions were performed through SWISS-Model (structure prediction) and ClusPro (molecular docking). Molecular investigation of the present LCA family identified a novel homozygous missense mutation p.Asp306Val in GUCY2D gene (NM_000180.3:c.917A>T). In silico structural modelling and interaction studies predicted significant changes in protein folding and interacting residues. The present molecular genetic study further extends the mutational spectrum of GUCY2D in LCA, and its genetic heterogeneity in the Pakistani population. The findings of the computational studies on protein structure and interaction profile predicted pathogenic consequences of p.Asp306Val on GUCY2D function.
- Published
- 2021
- Full Text
- View/download PDF
50. RelSim: An integrated method to identify disease genes using gene expression profiles and PPIN based similarity measure.
- Author
-
Maji, Pradipta, Shah, Ekta, and Paul, Sushmita
- Subjects
- *
GENE expression , *FUNCTIONAL genomics , *PROTEIN-protein interactions , *GENETICS of colon cancer , *FEATURE selection - Abstract
One of the important problems in functional genomics is how to select the disease genes. In this regard, the paper presents a new gene selection algorithm, termed as RelSim, to identify disease genes. It integrates judiciously the information of gene expression profiles and protein-protein interaction networks. A new similarity measure is introduced to compute the functional similarity between two genes. It is based on the information of protein-protein interaction networks. The new similarity measure offers an efficient way to calculate the functional similarity between two genes. The proposed algorithm selects a set of genes as disease genes, considering both microarray and protein-protein interaction data, by maximizing the relevance and functional similarity of the selected genes. While gene expression profiles are used to identify differentially expressed genes, the protein-protein interaction networks help to compute the functional similarity among genes. The performance of the proposed algorithm, along with a comparison with other related methods, is demonstrated on several colon cancer data sets. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.