Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 May 30.
Article in English | MEDLINE | ID: mdl-37398316

ABSTRACT

Quantifying behavior and relating it to underlying biological states is of paramount importance in many life science fields. Although barriers to recording postural data have been reduced by progress in deep-learning-based computer vision tools for keypoint tracking, extracting specific behaviors from this data remains challenging. Manual behavior coding, the present gold standard, is labor-intensive and subject to intra- and inter-observer variability. Automatic methods are stymied by the difficulty of explicitly defining complex behaviors, even ones which appear obvious to the human eye. Here, we demonstrate an effective technique for detecting one such behavior, a form of locomotion characterized by stereotyped spinning, termed 'circling'. Though circling has an extensive history as a behavioral marker, at present there exists no standard automated detection method. Accordingly, we developed a technique to identify instances of the behavior by applying simple postprocessing to markerless keypoint data from videos of freely-exploring (Cib2-/-;Cib3-/-) mutant mice, a strain we previously found to exhibit circling. Our technique agrees with human consensus at the same level as do individual observers, and it achieves >90% accuracy in discriminating videos of wild type mice from videos of mutants. As using this technique requires no experience writing or modifying code, it also provides a convenient, noninvasive, quantitative tool for analyzing circling mouse models. Additionally, as our approach was agnostic to the underlying behavior, these results support the feasibility of algorithmically detecting specific, research-relevant behaviors using readily-interpretable parameters tuned on the basis of human consensus.

2.
Arab J Urol ; 18(4): 252-256, 2020 Aug 13.
Article in English | MEDLINE | ID: mdl-33312737

ABSTRACT

OBJECTIVE: To compare a modified technique using the Dormia basket vs Stone Cone for stone entrapment to avoid proximal stone migration during ureteroscopic pneumatic lithotripsy of ureteric stones. PATIENTS AND METHODS: Our study included all patients with ureteric stones of <15 mm who underwent ureteroscopic pneumatic lithotripsy from January 2015 to September 2018. The study had two arms that were conducted over two consecutive periods; the first included 72 patients in whom we used the Stone Cone (Group 1) and the second included 86 patients in whom we started to use a Dormia basket with a modification (Group 2) to guard against proximal stone migration. RESULTS: Both groups were comparable for gender, age, and stone characteristics. Lower ureteric stones were the most prevalent as they represented 62.5% and 60.5% in groups 1 and 2, respectively; while upper ureteric stones were respectively found in 16.7% and 17.4%. Chemical stone analysis revealed that calcium oxalate stones were most predominant accounting for 51.3% and 51.1% in groups 1 and 2, respectively. Most of the stones were radio-opaque stones representing 57% and 58.1% in groups 1 and 2, respectively. There was a significant difference in operative time, with a mean (SD) operative time was 50.9 (11.2) in Group 1 vs 58.3 (12.4) min in Group 2 (P < 0.001). The success rate, defined as no retropulsion of stone fragments, was 97.7% in Group 2 vs 91.7% in Group 1 (P < 0.01). Complications were minor and comparable between the groups. There was no difference in hospital stay between the groups, but the cost assessment favoured Group 2. CONCLUSION: We found that our modified-basket stone entrapment technique compared favourably with the Stone Cone to guard against stone retropulsion during ureteroscopic pneumatic lithotripsy. Our modification to the basket was found to be feasible, efficient, safe, reproducible and cost-effective in preventing proximal stone migration. This procedure is particularly suitable in cost-limited environments.

4.
Clin Genet ; 93(3): 682-686, 2018 03.
Article in English | MEDLINE | ID: mdl-28940338

ABSTRACT

Congenital cataract (CC) is clinically and genetically highly heterogeneous. Here, we enrolled a consanguineous kindred (LUCC15) from Pakistan, with 3 affected individuals suffering with CC. Exome sequencing revealed a transition mutation [c.149 T > C; p.(Ile50Thr)] in INPP5K. Inositol polyphosphate-5-phosphatase K, encoded by INPP5K, is involved in dephosphorylation of phosphatidylinositol (PtdIns) 4,5-bisphosphate, and PtdIns 3,4,5-trisphosphate. Recently, pathogenic variants in INPP5K have been reported in families with congenital muscular dystrophies, intellectual disability, and cataract. In our family LUCC15, mild to moderate intellectual disability along with speech impairment was observed in 2 affected individuals. Magnetic resonance imaging of brain and muscles tissues did not reveal any cerebellar or muscular atrophy. However, electromyography of both upper and lower limbs revealed irritable myopathy. Comparison of clinical phenotype of all the known affected individuals, including LUCC15 family, homozygous for INPP5K alleles revealed reduced penetrance of muscular dystrophy and intellectual disability. Similarly, skeletal muscle abnormalities were highly variable among inpp5ka zebrafish mutants analyzed in this study. These phenotypic variabilities may be due to epigenetic factors and/or genetic modifiers.


Subject(s)
Cataract/congenital , Genes, Recessive , Genetic Variation , Phosphoric Monoester Hydrolases/genetics , Adolescent , Animals , Cataract/genetics , Child , Child, Preschool , Computational Biology/methods , Consanguinity , Female , Humans , Infant , Loss of Function Mutation , Magnetic Resonance Imaging , Male , Pakistan , Pedigree , Phenotype , Phosphoric Monoester Hydrolases/chemistry , Exome Sequencing , Zebrafish
5.
Mol Psychiatry ; 22(11): 1604-1614, 2017 11.
Article in English | MEDLINE | ID: mdl-27457812

ABSTRACT

Intellectual disability (ID) is a clinically and genetically heterogeneous disorder, affecting 1-3% of the general population. Although research into the genetic causes of ID has recently gained momentum, identification of pathogenic mutations that cause autosomal recessive ID (ARID) has lagged behind, predominantly due to non-availability of sizeable families. Here we present the results of exome sequencing in 121 large consanguineous Pakistani ID families. In 60 families, we identified homozygous or compound heterozygous DNA variants in a single gene, 30 affecting reported ID genes and 30 affecting novel candidate ID genes. Potential pathogenicity of these alleles was supported by co-segregation with the phenotype, low frequency in control populations and the application of stringent bioinformatics analyses. In another eight families segregation of multiple pathogenic variants was observed, affecting 19 genes that were either known or are novel candidates for ID. Transcriptome profiles of normal human brain tissues showed that the novel candidate ID genes formed a network significantly enriched for transcriptional co-expression (P<0.0001) in the frontal cortex during fetal development and in the temporal-parietal and sub-cortex during infancy through adulthood. In addition, proteins encoded by 12 novel ID genes directly interact with previously reported ID proteins in six known pathways essential for cognitive function (P<0.0001). These results suggest that disruptions of temporal parietal and sub-cortical neurogenesis during infancy are critical to the pathophysiology of ID. These findings further expand the existing repertoire of genes involved in ARID, and provide new insights into the molecular mechanisms and the transcriptome map of ID.


Subject(s)
Intellectual Disability/genetics , Alleles , Consanguinity , Exome/genetics , Family , Gene Frequency/genetics , Genetic Association Studies/methods , Humans , Mutation , Pakistan , Pedigree , Exome Sequencing/methods
6.
Clin Genet ; 90(1): 90-5, 2016 07.
Article in English | MEDLINE | ID: mdl-26572954

ABSTRACT

Skeletal dysplasias (SDs) are highly heterogeneous disorders composed of 40 clinical sub-types that are part of 456 well-delineated syndromes in humans. Here, we enrolled consanguineous kindred from a remote area of Sindh province of Pakistan, with 14 affected individuals suffering with short stature, kyphoscoliosis, joint dislocations, clubfoot, heart valve anomalies and progressive bilateral mixed hearing loss. To identify pathogenic variants in this family, whole exome sequencing (WES) was performed in one affected and one normal individual, which revealed a novel transversion mutation (c.802G>T; p.Glu268*) in CHST3 associated with the phenotype. CHST3 encodes a chondroitin 6-O-sulfotransferase-1 (C6ST-1) enzyme that is essential for the sulfation of proteoglycans found in cartilages. Previously, mutations in CHST3 have largely been reported in sporadic cases of SD, primarily with severe spinal abnormalities, joint dislocations, joint contractures, and clubfoot. Clinical and radiological examination of the affected individuals in this family provides new insights into phenotypic spectrum of CHST3 alleles and disease progression with age.


Subject(s)
Alleles , Hearing Loss, Bilateral/genetics , Heart Valve Diseases/genetics , Mutation , Osteochondrodysplasias/congenital , Sulfotransferases/genetics , Adult , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Consanguinity , Disease Progression , Exome , Female , Gene Expression , Hearing Loss, Bilateral/complications , Hearing Loss, Bilateral/diagnostic imaging , Hearing Loss, Bilateral/pathology , Heart Valve Diseases/complications , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Molecular , Osteochondrodysplasias/complications , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Pedigree , Phenotype , Carbohydrate Sulfotransferases
8.
Asian J Neurosurg ; 8(3): 147-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24403957

ABSTRACT

AIM: In this study, we were assessing the outcome of Endoscopic Third Ventriculostomy (ETV) in infants below six months of age in cases of congenital obstructive hydrocephalus. MATERIALS AND METHODS: The study was done prospectively on eight cases of obstructive hydrocephalus in infants younger than six months of age to assess the success rate of ETV as a primary treatment for hydrocephalus in this age group; in cases of evident failure, a ventriculo-peritoneal (VP) shunt was applied. RESULTS: Despite eliminating the factors suggested as causes of ETV failure in infants below six months; the type, as with the communicating hydrocephalus, the thickness of the third ventricular floor, history of previous intracranial hemorrhage or central nervous system infection, still the success rate did not exceed 12.5%. CONCLUSIONS: The complication rate following ETV was low in comparison to the high frequency (20-80%) and seriousness of the possible postoperative complications following VP shunt with a significant decrease in the quality of patients' lives. Hence the decision-making as well as the parental counselling were in a trial to estimate the ETV success or the need to perform a shunt in the treatment of obstructive hydrocephalus.

10.
Clin Genet ; 76(3): 270-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19650862

ABSTRACT

Autosomal recessive nonsyndromic hearing impairment (ARNSHI) segregating in three unrelated, large consanguineous Pakistani families (PKDF528, PKDF859 and PKDF326) is linked to markers on chromosome 12q14.2-q15. This novel locus is designated DFNB74. Maximum two-point limit of detection (LOD) scores of 5.6, 5.7 and 2.6 were estimated for markers D12S313,D12S83 and D12S75 at theta = 0 for recessive deafness segregating in these three families. Haplotype analyses identified a critical linkage interval of 5.35 cM (5.36 Mb) defined by D12S329 at 74.58 cM and D12S313 at 79.93 cM. DFNB74 is the second ARNSHI locus mapped to chromosome 12, but the physical intervals do not overlap with one another. A locus contributing to the early onset, rapidly progressing hearing loss of A/J mice (ahl4, age-related hearing loss 4) was reported to map to chromosome 10 in a region of conserved synteny to DFNB74, suggesting that ahl4 and DFNB74 may be due to mutations of the same gene in these two species.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Genes, Recessive , Genetic Loci , Hearing Loss/genetics , Audiometry, Pure-Tone , Chromosome Segregation/genetics , Family , Female , Humans , Lod Score , Male , Pedigree
11.
Clin Genet ; 75(3): 237-43, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19250381

ABSTRACT

Mutations in OTOF, encoding otoferlin, cause non-syndromic recessive hearing loss. The goal of our study was to define the identities and frequencies of OTOF mutations in a model population. We screened a cohort of 557 large consanguineous Pakistani families segregating recessive, severe-to-profound, prelingual-onset deafness for linkage to DFNB9. There were 13 families segregating deafness consistent with linkage to markers for DFNB9. We analyzed the genomic nucleotide sequence of OTOF and detected probable pathogenic sequence variants among all 13 families. These include the previously reported nonsense mutation p.R708X and 10 novel variants: 3 nonsense mutations (p.R425X, p.W536X, and p.Y1603X), 1 frameshift (c.1103_1104delinsC), 1 single amino acid deletion (p.E766del) and 5 missense substitutions of conserved residues (p.L573R, p.A1090E, p.E1733K, p.R1856Q and p.R1939W). OTOF mutations thus account for deafness in 13 (2.3%) of 557 Pakistani families. This overall prevalence is similar, but the mutation spectrum is different from those for Western populations. In addition, we demonstrate the existence of an alternative splice isoform of OTOF expressed in the human cochlea. This isoform must be required for human hearing because it encodes a unique alternative C-terminus affected by some DFNB9 mutations.


Subject(s)
Deafness/genetics , Gene Frequency/genetics , Membrane Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Cochlea/metabolism , Exons , Genes, Recessive , Genetic Variation , Humans , Membrane Proteins/metabolism , Molecular Sequence Data , Mutation , Pakistan , Pedigree , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Clin Genet ; 75(1): 86-91, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18505454

ABSTRACT

Usher syndrome (USH) is a hereditary disorder associated with sensorineural hearing impairment, progressive loss of vision attributable to retinitis pigmentosa (RP) and variable vestibular function. Three clinical types have been described with type I (USH1) being the most severe. To date, six USH1 loci have been reported. We ascertained two large Pakistani consanguineous families segregating profound hearing loss, vestibular dysfunction, and RP, the defining features of USH1. In these families, we excluded linkage of USH to the 11 known USH loci and subsequently performed a genome-wide linkage screen. We found a novel USH1 locus designated USH1H that mapped to chromosome 15q22-23 in a 4.92-cM interval. This locus overlaps the non-syndromic deafness locus DFNB48 raising the possibility that the two disorders may be caused by allelic mutations.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Usher Syndromes/genetics , Adolescent , Adult , Aged , Child , Chromosome Mapping , Female , Genotype , Humans , Male , Middle Aged , Mutation , Pedigree , Young Adult
13.
Clin Genet ; 72(6): 546-50, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17877751

ABSTRACT

Non-syndromic deafness is genetically heterogeneous. We previously reported that mutations of transmembrane channel-like gene 1 (TMC1) cause non-syndromic recessive deafness at the DFNB7/B11 locus on chromosome 9q13-q21 in nine Pakistani families. The goal of this study was to define the identities, origins and frequencies of TMC1 mutations in an expanded cohort of 557 large Pakistani families segregating recessive deafness. We screened affected family members for homozygosity at short-tandem repeats flanking known autosomal recessive (DFNB) deafness loci, followed by TMC1 sequence analysis in families segregating deafness linked to DFNB7/B11. We identified 10 new families segregating DFNB7/B11 deafness and TMC1 mutations, including three novel alleles. Overall, 9 different TMC1 mutations account for deafness in 19 (3.4%) of the 557 Pakistani families. A single mutation, p.R34X, causes deafness in 10 (1.8%) of the families. Genotype analysis of p.R34X-linked markers indicates that it arose from a common founder. We also detected p.R34X among normal control samples of African-American and northern European origins, raising the possibility that p.R34X and other mutations of TMC1 are prevalent contributors to the genetic load of deafness across a variety of populations and continents.


Subject(s)
Deafness/genetics , Membrane Proteins/genetics , Mutation , Amino Acid Sequence , Chromosomes, Human, Pair 9/genetics , Codon, Nonsense , Female , Gene Frequency , Genes, Recessive , Humans , Male , Molecular Sequence Data , Pakistan , Pedigree , Sequence Homology, Amino Acid
14.
J Med Genet ; 43(8): 634-40, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16459341

ABSTRACT

BACKGROUND: Approximately half the cases of prelingual hearing loss are caused by genetic factors. Identification of genes causing deafness is a crucial first step in understanding the normal function of these genes in the auditory system. Recently, a mutant allele of Tmhs was reported to be associated with deafness and circling behaviour in the hurry-scurry mouse. Tmhs encodes a predicted tetraspan protein of unknown function, which is expressed in inner ear hair cells. The human homologue of Tmhs is located on chromosome 6p. OBJECTIVE: To determine the cause of deafness in four consanguineous families segregating recessive deafness linked to markers on chromosome 6p21.1-p22.3 defining a novel DFNB locus. RESULTS: A novel locus for non-syndromic deafness DFNB67 was mapped in an interval of approximately 28.51 cM on human chromosome 6p21.1-p22.3. DNA sequence analysis of TMHS revealed a homozygous frameshift mutation (246delC) and a missense mutation (Y127C) in affected individuals of two families segregating non-syndromic deafness, one of which showed significant evidence of linkage to markers in the DFNB67 interval. The localisation of mTMHS in developing mouse inner ear hair cells was refined and found to be expressed briefly from E16.5 to P3. CONCLUSIONS: These findings establish the importance of TMHS for normal sound transduction in humans.


Subject(s)
Genes, Recessive/genetics , Hearing Loss/genetics , Inheritance Patterns , Membrane Proteins/genetics , Mutation, Missense/genetics , Alleles , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 6/genetics , DNA Mutational Analysis , Epithelium/metabolism , Genetic Linkage , Genetic Markers , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/metabolism , Humans , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pedigree , Physical Chromosome Mapping , Sequence Alignment
16.
Clin Genet ; 63(6): 431-44, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12786748

ABSTRACT

Association of sensorineural deafness and progressive retinitis pigmentosa with and without a vestibular abnormality is the hallmark of Usher syndrome and involves at least 12 loci among three different clinical subtypes. Genes identified for the more commonly inherited loci are USH2A (encoding usherin), MYO7A (encoding myosin VIIa), CDH23 (encoding cadherin 23), PCDH15 (encoding protocadherin 15), USH1C (encoding harmonin), USH3A (encoding clarin 1), and USH1G (encoding SANS). Transcripts from all these genes are found in many tissues/cell types other than the inner ear and retina, but all are uniquely critical for retinal and cochlear cell function. Many of these protein products have been demonstrated to have direct interactions with each other and perform an essential role in stereocilia homeostasis.


Subject(s)
Hearing Loss, Sensorineural/genetics , Retinitis Pigmentosa/genetics , Animals , Extracellular Matrix Proteins/genetics , Humans , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Vestibule, Labyrinth/abnormalities
18.
Am J Hum Genet ; 69(1): 25-34, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11398101

ABSTRACT

Human chromosome 10q21-22 harbors USH1F in a region of conserved synteny to mouse chromosome 10. This region of mouse chromosome 10 contains Pcdh15, encoding a protocadherin gene that is mutated in ames waltzer and causes deafness and vestibular dysfunction. Here we report two mutations of protocadherin 15 (PCDH15) found in two families segregating Usher syndrome type 1F. A Northern blot probed with the PCDH15 cytoplasmic domain showed expression in the retina, consistent with its pathogenetic role in the retinitis pigmentosa associated with USH1F.


Subject(s)
Cadherins/genetics , Chromosomes, Human, Pair 10/genetics , Deafness/genetics , Mutation/genetics , Protein Precursors/genetics , Retinitis Pigmentosa/genetics , Aged , Alleles , Animals , Base Sequence , Cadherin Related Proteins , Cadherins/chemistry , DNA Mutational Analysis , Female , Haplotypes , Humans , Lod Score , Male , Mice , Mice, Mutant Strains , Middle Aged , Pakistan , Pedigree , Phenotype , Physical Chromosome Mapping , Protein Precursors/chemistry , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Syndrome
19.
Am J Hum Genet ; 68(1): 26-37, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11090341

ABSTRACT

Genes causing nonsyndromic autosomal recessive deafness (DFNB12) and deafness associated with retinitis pigmentosa and vestibular dysfunction (USH1D) were previously mapped to overlapping regions of chromosome 10q21-q22. Seven highly consanguineous families segregating nonsyndromic autosomal recessive deafness were analyzed to refine the DFNB12 locus. In a single family, a critical region was defined between D10S1694 and D10S1737, approximately 0.55 cM apart. Eighteen candidate genes in the region were sequenced. Mutations in a novel cadherin-like gene, CDH23, were found both in families with DFNB12 and in families with USH1D. Six missense mutations were found in five families with DFNB12, and two nonsense and two frameshift mutations were found in four families with USH1D. A northern blot analysis of CDH23 showed a 9.5-kb transcript expressed primarily in the retina. CDH23 is also expressed in the cochlea, as is demonstrated by polymerase chain reaction amplification from cochlear cDNA.


Subject(s)
Alleles , Cadherins/genetics , Deafness/genetics , Genes, Recessive/genetics , Hearing Loss, Sensorineural/genetics , Mutation/genetics , Retinitis Pigmentosa/genetics , Amino Acid Sequence , Base Sequence , Cadherin Related Proteins , Chromosome Mapping , Consanguinity , DNA Mutational Analysis , DNA Primers , Exons/genetics , Female , Gene Frequency/genetics , Humans , Introns/genetics , Lod Score , Male , Pedigree , RNA, Messenger/analysis , RNA, Messenger/genetics , Syndrome
20.
Nat Genet ; 26(4): 431-4, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101839

ABSTRACT

More than 50% of severe childhood deafness is genetically determined, approximately 70% of which occurs without other abnormalities and is thus termed nonsyndromic. So far, 30 nonsyndromic recessive deafness loci have been mapped and the defective genes at 6 loci, DFNB1, DFNB2, DFNB3, DFNB4, DFNB9 and DNFB21, have been identified, encoding connexin-26 (ref. 3), myosin VIIA (ref. 4), myosin XV (ref. 5), pendrin, otoferlin and alpha-tectorin, respectively. Here we map a new recessive nonsyndromic deafness locus, DFNB26, to a 1.5-cM interval of chromosome 4q31 in a consanguineous Pakistani family. A maximum lod score of 8.10 at theta=0 was obtained with D4S1610 when only the 8 affected individuals in this family were included in the calculation. There are seven unaffected family members who are also homozygous for the DFNB26-linked haplotype and thus are non-penetrant. A dominant modifier, DFNM1, that suppresses deafness in the 7 nonpenetrant individuals was mapped to a 5.6-cM region on chromosome 1q24 with a lod score of 4.31 at theta=0 for D1S2815.


Subject(s)
Deafness/genetics , Chromosome Mapping , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 4/genetics , Connexin 26 , Connexins , Consanguinity , Female , Genes, Dominant , Genes, Recessive , Haplotypes , Humans , Lod Score , Male , Microsatellite Repeats , Pedigree , Suppression, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...