Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 143(3): 311-329, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38459354

RESUMEN

Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modeling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modeling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.


Asunto(s)
Sordera , Mutación Missense , Linaje , Receptores de Superficie Celular , Estereocilios , Animales , Femenino , Humanos , Masculino , Sordera/genética , Secuenciación del Exoma , Genes Recesivos , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Modelos Moleculares , Receptores de Superficie Celular/genética , Estereocilios/metabolismo , Estereocilios/patología , Estereocilios/genética
2.
Toxicol Appl Pharmacol ; 489: 117008, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38908719

RESUMEN

The current study aimed to determine the safety and efficacy of Coag-A through in vivo analysis in CFA induced mice model. Treatment of CFA induced arthritis in mice with Coagulansin-A (10 mg/kg i.p. daily for 28 days), a withanolide obtained from Withania coagulans, as well as standard drug treatment with Dexamethasone (5 mg/kg i.p) was provided. The effect of Coag-A on body weight, relative organ weight, hematology, serum biochemistry, survival rate, oxidative stress markers, and antioxidant enzymes was evaluated. The liver and kidney histopathology were also assessed to ascertain its safety profile. Treatment of arthritic mice with Coag-A considerably improved body weight, relative organ weight of liver, kidney, and spleen, ameliorated hematology and serum biochemistry, and increased survival and antioxidant potential. Coag-A was found to be safer with fewer adverse effects showing hepato-protective, nephroprotective, and anti-inflammatory effect. It also significantly (p < 0.001) improved histopathology of CFA-induced mice when compared with Dexa. In conclusion, compared to dexamethasone, Coag-A has demonstrated a greater therapeutic benefit and fewer side effects in the treatment of arthritis against the CFA-induced model.

3.
J Med Genet ; 60(4): 352-358, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36002240

RESUMEN

BACKGROUND: Dystonia involves repetitive movements and muscle contractions leading to abnormal postures. We investigated patients in two families, DYAF11 and M, exhibiting dystonic or involuntary movement disorders. METHODS: Clinical investigations were performed for all patients. Genetic analyses included genome-wide linkage analysis and exome sequencing followed by Sanger sequencing validation. MRM2-specific transcripts were analysed from participants' blood samples in Family DYAF11 after cloning of gene-specific cDNA. RESULTS: Four affected siblings in Family DYAF11 had progressive dystonic features. Two patients in Family M exhibited a neurodevelopmental disorder accompanied by involuntary movements. In Family DYAF11, linkage was detected to the telomere at chromosome 7p22.3, spanning <2 Mb. Exome sequencing identified a donor splice-site variant, c.8+1G>T in MRM2, which segregated with the phenotype, corresponding to the linkage data since all affected individuals were homozygous while the obligate unaffected carriers were heterozygous for the variant. In the MRM2 c.8+1G>T allele, an aberrant alternative acceptor splice-site located within exon 2 was used in a subset of the transcripts, creating a frameshift in the open reading frame. Exome sequencing in Family M revealed a rare missense variant c.242C>T, p.(Ala81Val), which affected a conserved amino acid. CONCLUSIONS: Our results expand the clinical and allelic spectrum of MRM2 variants. Previously, these descriptions were based on observations in a single patient, diagnosed with mitochondrial DNA depletion syndrome 17, in whom movement disorder was accompanied by recurrent strokes and epilepsy. We also demonstrate a subset of correctly spliced tt-ag MRM2 transcripts, raising the possibility to develop treatment by understanding the disease mechanism.


Asunto(s)
Mutación del Sistema de Lectura , Sitios de Empalme de ARN , Humanos , Mutación , Fenotipo , Exones , Síndrome , Linaje
4.
Inflammopharmacology ; 32(2): 1225-1238, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38411787

RESUMEN

The current work was designed to evaluate the anti-inflammatory and anti-arthritic potential of Coagulansin-A (Coag-A) using mouse macrophages and arthritic mice. In the LPS-induced RAW 264.7 cells, the effects of Coag-A on the release of nitric oxide (NO), reactive oxygen species (ROS), and pro-inflammatory cytokines were analyzed. In addition, the mediators involved in the nuclear factor-kappa B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways were evaluated by the RT-qPCR and western blotting. Coag-A did not show significant cytotoxicity in the RAW 264.7 cells in the tested concentration range (1-100 µM). Coag-A significantly inhibited the production of NO, ROS, and key pro-inflammatory cytokines. The anti-inflammatory effects of Coag-A might be through inhibiting the NF-κB pathway and activating the Nrf2 pathway. In the arthritic mouse models, behavioral studies and radiological and histological analyses were performed. We found that the i.p. injection of Coag-A dose-dependently (1-10 mg/kg) reduced the Carrageenan-induced acute inflammation in the mice. In Complete Freund's Reagent-induced arthritic mouse model, Coag-A (10 mg/kg) showed significant anti-inflammatory and anti-arthritic effects in terms of the arthritic index, hematological parameters, and synovium inflammation. After the Coag-A treatment, the bone and tissue damage was ameliorated significantly in the arthritic mice. Moreover, immunohistochemistry of mouse paw tissues revealed a significant reduction in the expression of pro-inflammatory cytokines in the NF-κB pathway, confirming Coag-A's therapeutic potential and mechanism.


Asunto(s)
Factor 2 Relacionado con NF-E2 , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Antiinflamatorios/uso terapéutico , Inflamación/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología
5.
BMC Musculoskelet Disord ; 24(1): 735, 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37710205

RESUMEN

BACKGROUND: Multicentric osteolysis nodulosis and arthropathy (MONA) is a rare autosomal recessive disorder characterized by marked progressive bone loss and joint destruction resulting in skeletal deformities. MONA is caused by MMP2 deficiency. Here we report clinical and molecular analyses of four patients in two families from Pakistan and Finland. METHODS: Clinical analyses including radiography were completed and blood samples were collected. The extracted DNA was subjected to whole-exome analysis or target gene sequencing. Segregation analyses were performed in the nuclear pedigree. Pathogenicity prediction scores for the selected variants and conservation analyses of affected amino acids were observed. RESULTS: The phenotype in the four affected individuals was consistent with multicentric osteolysis or MONA, as the patients had multiple affected joints, osteolysis of hands and feet, immobility of knee joint and progressive bone loss. Long-term follow up of the patients revealed the progression of the disease. We found a novel MMP2 c.1336 + 2T > G homozygous splice donor variant segregating with the phenotype in the Pakistani family while a MMP2 missense variant c.1188 C > A, p.(Ser396Arg) was homozygous in both Finnish patients. In-silico analysis predicted that the splicing variant may eventually introduce a premature stop codon in MMP2. Molecular modeling for the p.(Ser396Arg) variant suggested that the change may disturb MMP2 collagen-binding region. CONCLUSION: Our findings expand the genetic spectrum of Multicentric osteolysis nodulosis and arthropathy. We also suggest that the age of onset of this disorder may vary from childhood up to late adolescence and that a significant degree of intrafamilial variability may be present.


Asunto(s)
Síndrome de Hajdu-Cheney , Artropatías , Osteólisis , Adolescente , Humanos , Niño , Metaloproteinasa 2 de la Matriz , Artropatías/diagnóstico por imagen , Artropatías/genética , Osteólisis/diagnóstico por imagen , Osteólisis/genética
6.
Hum Genet ; 141(3-4): 633-648, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34308486

RESUMEN

Approximately 14.5 million Pakistani individuals have a hearing loss and half of these cases may be due to genetic causes. Though significant progress has been made in uncovering genetic variants for recessively inherited nonsyndromic deafness, Pendred syndrome, and Usher syndromes, the same is not true for dominantly inherited hearing loss, most syndromic cases and deafness with complex inheritance patterns. Variants of 57 genes have been reported to cause nonsyndromic recessive deafness in Pakistan, though most are rare. Variants of just five genes GJB2, HGF, MYO7A, SLC26A4, and TMC1 together explain 57% of profound deafness while those of GJB2, MYO15A, OTOF, SLC26A4, TMC1, and TMPRSS3 account for 47% of moderate to severe hearing loss. In contrast, although variants of at least 39 genes have been implicated in different deafness syndromes, their prevalence in the population and the spectrum of mutations have not been explored. Furthermore, research on genetics of deafness has mostly focused on individuals from the Punjab province and needs to be extended to other regions of Pakistan. Identifying the genes and their variants causing deafness in all ethnic groups is important as it will pinpoint rare as well as recurrent mutations. This information may ultimately help in offering genetic counseling and future treatments.


Asunto(s)
Sordera , Pérdida Auditiva , Conexina 26/genética , Conexinas/genética , Sordera/genética , Pérdida Auditiva/genética , Humanos , Proteínas de la Membrana/genética , Biología Molecular , Mutación , Proteínas de Neoplasias/genética , Pakistán/epidemiología , Serina Endopeptidasas/genética
7.
Hum Genet ; 141(3-4): 805-819, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34338890

RESUMEN

Hearing loss and impaired fertility are common human disorders each with multiple genetic causes. Sometimes deafness and impaired fertility, which are the hallmarks of Perrault syndrome, co-occur in a person. Perrault syndrome is inherited as an autosomal recessive disorder characterized by bilateral mild to severe childhood sensorineural hearing loss with variable age of onset in both sexes and ovarian dysfunction in females who have a 46, XX karyotype. Since the initial clinical description of Perrault syndrome 70 years ago, the phenotype of some subjects may additionally involve developmental delay, intellectual deficit and other neurological disabilities, which can vary in severity in part dependent upon the genetic variants and the gene involved. Here, we review the molecular genetics and clinical phenotype of Perrault syndrome and focus on supporting evidence for the eight genes (CLPP, ERAL1, GGPS1, HARS2, HSD17B4, LARS2, RMND1, TWNK) associated with Perrault syndrome. Variants of these eight genes only account for approximately half of the individuals with clinical features of Perrault syndrome where the molecular genetic base remains under investigation. Additional environmental etiologies and novel Perrault disease-associated genes remain to be identified to account for unresolved cases. We also report a new genetic variant of CLPP, computational structural insight about CLPP and single cell RNAseq data for eight reported Perrault syndrome genes suggesting a common cellular pathophysiology for this disorder. Some unanswered questions are raised to kindle future research about Perrault syndrome.


Asunto(s)
Aminoacil-ARNt Sintetasas , Disgenesia Gonadal 46 XX , Pérdida Auditiva Sensorineural , Aminoacil-ARNt Sintetasas/genética , Proteínas de Ciclo Celular/genética , Niño , Femenino , Disgenesia Gonadal 46 XX/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Mutación , Linaje
8.
J Psychiatry Neurosci ; 47(6): E379-E390, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36318984

RESUMEN

BACKGROUND: Schizophrenia is characterized by hallucinations, delusions and disorganized behaviour. Recessive or X-linked transmissions are rarely described for common psychiatric disorders. We examined the genetics of psychosis to identify rare large-effect variants in patients with extreme schizophrenia. METHODS: We recruited 2 consanguineous families, each with patients affected by early-onset, severe, treatment-resistant schizophrenia. We performed exome sequencing for all participants. We checked variant rarity in public databases and with ethnically matched controls. We performed in silico analyses to assess the effects of the variants on proteins. RESULTS: Structured clinical evaluations supported diagnoses of schizophrenia in all patients and phenotypic absence in the unaffected individuals. Data analyses identified multiple variants. Only 1 variant per family was predicted as pathogenic by prediction tools. A homozygous c.649C > T:p.(Arg217Cys) variant in RGS3 and a hemizygous c.700A > G:p.(Thr234Ala) variant in IL1RAPL1 affected evolutionary conserved amino acid residues and were the most likely causes of phenotype in the patients of each family. Variants were ultra-rare in publicly available databases and absent from the DNA of 400 ethnically matched controls. RGS3 is implicated in modulating sensory behaviour in Caenorhabditis elegans. Variants of IL1RAPL1 are known to cause nonsyndromic X-linked intellectual disability with or without human behavioural dysfunction. LIMITATIONS: Each variant is unique to a particular family's patients, and findings may not be replicated. CONCLUSION: Our work suggests that some rare variants may be involved in causing inherited psychosis or schizophrenia. Variant-specific functional studies will elucidate the pathophysiology relevant to schizophrenias and motivate translation to personalized therapeutics.


Asunto(s)
Proteínas RGS , Esquizofrenia , Humanos , Esquizofrenia/genética , Exoma , Linaje , Mutación Missense , Transmisión Sináptica , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteínas RGS/genética
9.
BMC Psychiatry ; 22(1): 709, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36384485

RESUMEN

BACKGROUND: Psychiatric disorders are characterized by alteration in emotions, mood and behavior. Genetics is known to play a significant role in the development of psychiatric disorders. Genome-wide association studies have identified several loci associated with psychiatric illnesses. We hypothesize the existence of rare variants following Mendelian recessive mode of inheritance. These variants can be identified in families with multiple affected individuals born to unaffected consanguineous parents. METHODS: We visited psychiatric outpatient departments of multiple hospitals in Lahore, Pakistan. We focused on psychosis, as it can occur in several DSM disorders such as schizophrenia, dementia and bipolar disorder. After clinical diagnosis by an American trained psychiatrist, detailed clinical assessments using Diagnostic Interview for Genetic Studies (DIGS), Diagnostic Interview for Psychosis and Affective Disorders (DI-PAD), Positive and Negative Syndrome Scale (PANSS), Hamilton Depression and Anxiety Rating Scale (HAM-D; HAM-A) were administered to all willing affected and unaffected participants. RESULTS: We identified eight pedigrees with two or more psychotic individuals in each family. Clinical diagnoses determined by their psychiatrists included ten individuals with schizophrenia; four individuals with psychosis and bipolar disorder; and two patients with "unspecified psychosis." The rating instruments rigorously confirmed the diagnosis of psychosis in the affected patients from the six families as well as the absence of psychotic disorders in unaffected individuals from the six families. We obtained DNA samples from willing members of all eight families for future genetic analyses. CONCLUSION: Our research highlights an alternative approach to discovery of rare recessively inherited genetic variants causing psychiatric disorders that have remained unidentified to date. These findings could illuminate underlying biological mechanisms leading toward development of targeted therapies in future.


Asunto(s)
Trastornos Mentales , Trastornos Psicóticos , Esquizofrenia , Humanos , Consanguinidad , Estudio de Asociación del Genoma Completo , Trastornos Mentales/genética , Trastornos Psicóticos/diagnóstico , Trastornos Psicóticos/genética , Esquizofrenia/diagnóstico
10.
J Med Genet ; 58(5): 351-356, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32591345

RESUMEN

BACKGROUND: Studies exploring molecular mechanisms underlying congenital skeletal disorders have revealed novel regulators of skeletal homeostasis and shown protein glycosylation to play an important role. OBJECTIVE: To identify the genetic cause of rhizomelic skeletal dysplasia in a consanguineous Pakistani family. METHODS: Clinical investigations were carried out for four affected individuals in the recruited family. Whole genome sequencing (WGS) was completed using DNA from two affected and two unaffected individuals from the family. Sequencing data were processed, filtered and analysed. In silico analyses were performed to predict the effects of the candidate variant on the protein structure and function. Small interfering RNAs (siRNAs) were used to study the effect of Gnpnat1 gene knockdown in primary rat chondrocytes. RESULTS: The patients presented with short stature due to extreme shortening of the proximal segments of the limbs. Radiographs of one individual showed hip dysplasia and severe platyspondyly. WGS data analyses identified a homozygous missense variant c.226G>A; p.(Glu76Lys) in GNPNAT1, segregating with the disease. Glucosamine 6-phosphate N-acetyltransferase, encoded by the highly conserved gene GNPNAT1, is one of the enzymes required for synthesis of uridine diphosphate N-acetylglucosamine, which participates in protein glycosylation. Knockdown of Gnpnat1 by siRNAs decreased cellular proliferation and expression of chondrocyte differentiation markers collagen type 2 and alkaline phosphatase, indicating that Gnpnat1 is important for growth plate chondrocyte proliferation and differentiation. CONCLUSIONS: This study describes a novel severe skeletal dysplasia associated with a biallelic, variant in GNPNAT1. Our data suggest that GNPNAT1 is important for growth plate chondrogenesis.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Fémur/anomalías , Glucosamina 6-Fosfato N-Acetiltransferasa/genética , Húmero/anomalías , Adulto , Anciano , Anciano de 80 o más Años , Animales , Enfermedades del Desarrollo Óseo/diagnóstico por imagen , Enfermedades del Desarrollo Óseo/patología , Células Cultivadas , Consanguinidad , Femenino , Fémur/diagnóstico por imagen , Fémur/patología , Homocigoto , Humanos , Húmero/diagnóstico por imagen , Húmero/patología , Masculino , Persona de Mediana Edad , Linaje , Radiografía , Ratas Sprague-Dawley
11.
J Med Genet ; 58(4): 237-246, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32439809

RESUMEN

BACKGROUND: Intellectual disability syndromes (IDSs) with or without developmental delays affect up to 3% of the world population. We sought to clinically and genetically characterise a novel IDS segregating in five unrelated consanguineous families. METHODS: Clinical analyses were performed for eight patients with intellectual disability (ID). Whole-exome sequencing for selected participants followed by Sanger sequencing for all available family members was completed. Identity-by-descent (IBD) mapping was carried out for patients in two Egyptian families harbouring an identical variant. RNA was extracted from blood cells of Turkish participants, followed by cDNA synthesis and real-time PCR for TTC5. RESULTS: Phenotype comparisons of patients revealed shared clinical features of moderate-to-severe ID, corpus callosum agenesis, mild ventriculomegaly, simplified gyral pattern, cerebral atrophy, delayed motor and verbal milestones and hypotonia, presenting with an IDS. Four novel homozygous variants in TTC5: c.629A>G;p.(Tyr210Cys), c.692C>T;p.(Ala231Val), c.787C>T;p.(Arg263Ter) and c.1883C>T;p.(Arg395Ter) were identified in the eight patients from participating families. IBD mapping revealed that c.787C>T;p.(Arg263Ter) is a founder variant in Egypt. Missense variants c.629A>G;p.(Tyr210Cys) and c.692C>T;p.(Ala231Val) disrupt highly conserved residues of TTC5 within the fifth and sixth tetratricopeptide repeat motifs which are required for p300 interaction, while the nonsense variants are predicted to decrease TTC5 expression. Functional analysis of variant c.1883C>T;p.(Arg395Ter) showed reduced TTC5 transcript levels in accordance with nonsense-mediated decay. CONCLUSION: Combining our clinical and molecular data with a recent case report, we identify the core and variable clinical features associated with TTC5 loss-of-function variants and reveal the requirement for TTC5 in human brain development and health.


Asunto(s)
Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Alelos , Niño , Preescolar , Discapacidades del Desarrollo/epidemiología , Discapacidades del Desarrollo/patología , Egipto/epidemiología , Exoma/genética , Femenino , Homocigoto , Humanos , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/patología , Masculino , Mutación/genética , Linaje , Fenotipo , Secuenciación del Exoma
12.
BMC Musculoskelet Disord ; 23(1): 818, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36042462

RESUMEN

BACKGROUND: Skeletal dysplasia is a heterogeneous group of disorders. Spondyloepiphyseal dysplasias comprise one subgroup. Deficiency of carbohydrate sulfotransferase 3 has been reported in a small number of patients with recessively inherited spondyloepiphyseal dysplasia with joint dislocation, short stature and scoliosis. We report here molecular and clinical findings of affected individuals in three consanguineous Pakistani families. Affected individuals in all three families had a uniform phenotype including severe short stature, multiple dislocated joints, progressive scoliosis and facial dysmorphism. METHODS: Clinical evaluation was done for three unrelated families. Radiological survey of bones was completed for patients from two of the families. Whole exome sequencing index patients from each family was performed followed by Sanger sequencing for validation of segregation of identified variants in respective families. In-silico analysis for determining pathogenicity of identified variants and conservation was done. RESULTS: Whole-exome sequencing revealed biallelic variants c.590 T > C;p.(Leu197Pro), c.603C > A;p.(Tyr201Ter) and c.661C > T;p.(Arg221Cys) in CHST3 (NM_004273.5) in the three families with eight, five and two affected individuals, respectively. Contrary to previous reports, affected individuals in none of the families exhibited a hearing loss. CONCLUSION: We describe genotypic and phenotypic findings of three unrelated families with spondyloepiphyseal dysplasia. Our study confirms phenotypic variability and adds to the genotypic spectrum of spondyloepiphyseal dysplasia.


Asunto(s)
Luxaciones Articulares , Osteocondrodisplasias , Escoliosis , Sulfotransferasas , Humanos , Mutación , Osteocondrodisplasias/congénito , Osteocondrodisplasias/diagnóstico por imagen , Osteocondrodisplasias/genética , Pakistán , Linaje , Fenotipo , Sulfotransferasas/genética , Carbohidrato Sulfotransferasas
13.
Int J Intell Syst ; 37(3): 2371-2392, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37520859

RESUMEN

The coronavirus of 2019 (COVID-19) was declared a global pandemic by World Health Organization in March 2020. Effective testing is crucial to slow the spread of the pandemic. Artificial intelligence and machine learning techniques can help COVID-19 detection using various clinical symptom data. While deep learning (DL) approach requiring centralized data is susceptible to a high risk of data privacy breaches, federated learning (FL) approach resting on decentralized data can preserve data privacy, a critical factor in the health domain. This paper reviews recent advances in applying DL and FL techniques for COVID-19 detection with a focus on the latter. A model FL implementation use case in health systems with a COVID-19 detection using chest X-ray image data sets is studied. We have also reviewed applications of previously published FL experiments for COVID-19 research to demonstrate the applicability of FL in tackling health research issues. Last, several challenges in FL implementation in the healthcare domain are discussed in terms of potential future work.

14.
Hum Mutat ; 42(10): 1321-1335, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34265170

RESUMEN

Hereditary deafness is clinically and genetically heterogeneous. We investigated deafness segregating as a recessive trait in two families. Audiological examinations revealed an asymmetric mild to profound hearing loss with childhood or adolescent onset. Exome sequencing of probands identified a homozygous c.475G>A;p.(Glu159Lys) variant of CLDN9 (NM_020982.4) in one family and a homozygous c.370_372dupATC;p.(Ile124dup) CLDN9 variant in an affected individual of a second family. Claudin 9 (CLDN9) is an integral membrane protein and constituent of epithelial bicellular tight junctions (TJs) that form semipermeable, paracellular barriers between inner ear perilymphatic and endolymphatic compartments. Computational structural modeling predicts that substitution of a lysine for glutamic acid p.(Glu159Lys) alters one of two cis-interactions between CLDN9 protomers. The p.(Ile124dup) variant is predicted to locally misfold CLDN9 and mCherry tagged p.(Ile124dup) CLDN9 is not targeted to the HeLa cell membrane. In situ hybridization shows that mouse Cldn9 expression increases from embryonic to postnatal development and persists in adult inner ears coinciding with prominent CLDN9 immunoreactivity in TJs of epithelia outlining the scala media. Together with the Cldn9 deaf mouse and a homozygous frameshift of CLDN9 previously associated with deafness, the two bi-allelic variants of CLDN9 described here point to CLDN9 as a bona fide human deafness gene.


Asunto(s)
Claudinas , Sordera , Adolescente , Animales , Niño , Claudinas/genética , Sordera/genética , Células HeLa , Homocigoto , Humanos , Ratones , Mutación , Linaje
15.
Hum Mutat ; 42(1): 89-101, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33252156

RESUMEN

Skeletal dysplasias are a heterogeneous group of disorders ranging from mild to lethal skeletal defects. We investigated two unrelated families with individuals presenting with a severe skeletal disorder. In family NMD02, affected individuals had a dysostosis multiplex-like skeletal dysplasia and severe short stature (<-8.5 SD). They manifested increasingly coarse facial features, protruding abdomens, and progressive skeletal changes, reminiscent of mucopolysaccharidosis. The patients gradually lost mobility and the two oldest affected individuals died in their twenties. The affected child in family ID01 had coarse facial features and severe skeletal dysplasia with clinical features similar to mucopolysaccharidosis. She had short stature, craniosynostosis, kyphoscoliosis, and hip-joint subluxation. She died at the age of 5 years. Whole-exome sequencing identified two homozygous variants c.133C>T; p.(Arg45Trp) and c.215dupA; p.(Tyr72Ter), respectively, in the two families, affecting an evolutionary conserved gene TMEM251 (NM_001098621.1). Immunofluorescence and confocal studies using human osteosarcoma cells indicated that TMEM251 is localized to the Golgi complex. However, p.Arg45Trp mutant TMEM251 protein was targeted less efficiently and the localization was punctate. Tmem251 knockdown by small interfering RNA induced dedifferentiation of rat primary chondrocytes. Our work implicates TMEM251 in the pathogenesis of a novel disorder and suggests its potential function in chondrocyte differentiation.


Asunto(s)
Enanismo , Proteínas de la Membrana , Osteocondrodisplasias , Animales , Femenino , Humanos , Ratas , Enanismo/genética , Secuenciación del Exoma , Homocigoto , Proteínas de la Membrana/genética , Osteocondrodisplasias/genética , Linaje
16.
BMC Musculoskelet Disord ; 22(1): 31, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407338

RESUMEN

BACKGROUND: Loss of function or gain of function variants of Filamin B (FLNB) cause recessive or dominant skeletal disorders respectively. Spondylocarpotarsal synostosis syndrome (SCT) is a rare autosomal recessive disorder characterized by short stature, fused vertebrae and fusion of carpal and tarsal bones. We present a novel FLNB homozygous pathogenic variant and present a carrier of the variant with short height. CASE PRESENTATION: We describe a family with five patients affected with skeletal malformations, short stature and vertebral deformities. Exome sequencing revealed a novel homozygous frameshift variant c.2911dupG p.(Ala971GlyfsTer122) in FLNB, segregating with the phenotype in the family. The variant was absent in public databases and 100 ethnically matched control chromosomes. One of the heterozygous carriers of the variant had short stature. CONCLUSION: Our report expands the genetic spectrum of FLNB pathogenic variants. It also indicates a need to assess the heights of other carriers of FLNB recessive variants to explore a possible role in idiopathic short stature.


Asunto(s)
Escoliosis , Sinostosis , Anomalías Múltiples , Filaminas/genética , Humanos , Vértebras Lumbares/anomalías , Enfermedades Musculoesqueléticas , Escoliosis/congénito , Escoliosis/diagnóstico por imagen , Escoliosis/genética , Sinostosis/diagnóstico por imagen , Sinostosis/genética , Vértebras Torácicas/anomalías , Vértebras Torácicas/diagnóstico por imagen
17.
BMC Musculoskelet Disord ; 22(1): 630, 2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34284742

RESUMEN

BACKGROUND: Skeletal dysplasia is a heterogeneous group of disorders resulting from different genetic variants in humans. The current study was designed to identify the genetic causes of skeletal dysplasia and short stature in two consanguineous families from Pakistan, both comprised of multiple affected individuals. Patients in one family had proportionate short stature with reduced head circumference while affected individuals in the other family had disproportionate short stature. METHODS: Clinical data were obtained and radiological examinations of the index patients were completed. Whole genome sequencing for probands from both families were performed followed by Sanger sequencing to confirm segregation of identified variants in the respective families. In-silico pathogenicity score prediction for identified variant and amino acid conservation analysis was completed. RESULTS: Whole Genome Sequencing identified a known biallelic variant c.6176_6189delGTCAGCTGCCGAAG; p.(Gln2060ArgfsTer48) in PCNT gene and a novel biallelic variant c.174delC; p.(Asp60ThrfsTer7) in RAB33B gene respectively in affected members of the two families. Clinical imaging revealed platyspondyly and varus deformity in the legs of the affected members in the first family. Radiographs indicated severe platyspondyly, genu valgus deformity of legs and pectus carinatum for the patients in the second family. CONCLUSION: In this study we report the phenotypes and genetic variants in two unrelated families with two distinct forms of skeletal dysplasia. This study strengthens the previous findings that patients harboring PCNT variants are phenotypically homogeneous and also extends the genotypic spectrum of RAB33B variants.


Asunto(s)
Osteocondrodisplasias , Consanguinidad , Humanos , Pakistán , Linaje , Fenotipo , Proteínas de Unión al GTP rab
18.
Hum Mol Genet ; 27(5): 780-798, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29293958

RESUMEN

The Cell Division-Cycle-14 gene encodes a dual-specificity phosphatase necessary in yeast for exit from mitosis. Numerous disparate roles of vertebrate Cell Division-Cycle-14 (CDC14A) have been proposed largely based on studies of cultured cancer cells in vitro. The in vivo functions of vertebrate CDC14A are largely unknown. We generated and analyzed mutations of zebrafish and mouse CDC14A, developed a computational structural model of human CDC14A protein and report four novel truncating and three missense alleles of CDC14A in human families segregating progressive, moderate-to-profound deafness. In five of these families segregating pathogenic variants of CDC14A, deaf males are infertile, while deaf females are fertile. Several recessive mutations of mouse Cdc14a, including a CRISPR/Cas9-edited phosphatase-dead p.C278S substitution, result in substantial perinatal lethality, but survivors recapitulate the human phenotype of deafness and male infertility. CDC14A protein localizes to inner ear hair cell kinocilia, basal bodies and sound-transducing stereocilia. Auditory hair cells of postnatal Cdc14a mutants develop normally, but subsequently degenerate causing deafness. Kinocilia of germ-line mutants of mouse and zebrafish have normal lengths, which does not recapitulate the published cdc14aa knockdown morphant phenotype of short kinocilia. In mutant male mice, degeneration of seminiferous tubules and spermiation defects result in low sperm count, and abnormal sperm motility and morphology. These findings for the first time define a new monogenic syndrome of deafness and male infertility revealing an absolute requirement in vivo of vertebrate CDC14A phosphatase activity for hearing and male fertility.


Asunto(s)
Pérdida Auditiva/genética , Infertilidad Masculina/genética , Monoéster Fosfórico Hidrolasas/genética , Proteínas Tirosina Fosfatasas/genética , Animales , Sistemas CRISPR-Cas , Femenino , Estudios de Asociación Genética , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Ratones Mutantes , Linaje , Monoéster Fosfórico Hidrolasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Testículo/fisiopatología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
19.
Clin Genet ; 97(1): 138-155, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31506927

RESUMEN

A variety of different signaling pathways are necessary for development and maintenance of the human auditory system. Normal hearing allows for the detection of soft sounds within the frequency range of 20 to 20 000 Hz, but more importantly to perceive the human voice frequency band of 250 to 6000 Hz. Loss of hearing is common, and is a clinically heterogeneous disorder that can be caused by environmental factors such as exposure to loud noise, infections and ototoxic drugs. In addition, variants of hundreds of genes have been reported to disrupt processes required for hearing. Noncoding regulatory variants and variants of additional genes necessary for hearing remain to be discovered as many individuals with inherited deafness are without a genetic diagnosis, despite the advent of whole exome sequencing. Here, we discuss in detail some of these deafness-causing variants of genes encoding a ligand or its receptor. Spotlighted in this review are three growth factor-receptor-pairs EDN3/EDNRB, HGF/MET and JAG/NOTCH, which individually are necessary for normal hearing. We also offer our perspective on unanswered questions, future challenges and potential opportunities for treatments emerging from molecular genetic and mechanistic studies of deafness due to these causes.


Asunto(s)
Sordera/genética , Endotelina-3/genética , Pérdida Auditiva Sensorineural/genética , Factor de Crecimiento de Hepatocito/genética , Receptor de Endotelina B/genética , Sordera/patología , Audición/genética , Audición/fisiología , Pérdida Auditiva Sensorineural/patología , Humanos , Proteína Jagged-1/genética , Proteínas Proto-Oncogénicas c-met/genética , Receptores Notch/genética
20.
J Pak Med Assoc ; 70(2): 208-212, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32063608

RESUMEN

OBJECTIVE: To determine the association of hepatocyte growth factor gene single nucleotide polymorphismsrs 5745718 and rs17427817 with primary angle closure glaucoma.. METHODS: This case-control study was conducted at the Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan, from August 2016 to September 2017. In this study seventy sporadic cases of primary angle closure glaucoma and sixty healthy controls were enrolled from different hospitals of the Lahore, Punjab. Blood samples were obtained from all the subjects. Genomic deoxyribonucleic acid was extracted by non-organic method. Two single nucleotide polymorphism (SNPs) rs5745718 and rs17427817 in Hepatocytes Growth Factor (HFG) gene were genotyped in patients and ethnically same healthy controls by using polymerase chain reaction restriction fragment length polymorphism (RFLP) assay. Data was analysed using SPSS (version20). RESULTS: Of the 130 subjects, 70(54%) were cases and 60(46%) controls. The mean age of the cases was 54±17 years (range: 13-85 years). The differences in genotype distribution were statistically significant for rs 5745718 (p=0.005 and p=0.009), while results were not significant for rs 17427817 (p=0.06 and p=0.09) between the cases and the controls. CONCLUSIONS: AC alleles were found to be protective while CC alleles were a risk factor for primary angle closure glaucoma in rs5745718 single nucleotide polymorphism.


Asunto(s)
Glaucoma de Ángulo Cerrado/genética , Factor de Crecimiento de Hepatocito/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Predisposición Genética a la Enfermedad , Glaucoma de Ángulo Cerrado/fisiopatología , Humanos , Presión Intraocular , Masculino , Persona de Mediana Edad , Pakistán , Polimorfismo Genético , Polimorfismo de Nucleótido Simple , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA