Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Firoz Kabir is active.

Publication


Featured researches published by Firoz Kabir.


Nature Communications | 2016

FOXE3 contributes to Peters anomaly through transcriptional regulation of an autophagy-associated protein termed DNAJB1.

Shahid Y. Khan; Shivakumar Vasanth; Firoz Kabir; John D. Gottsch; Arif O. Khan; Raghothama Chaerkady; Mei Chong W Lee; Carmen C. Leitch; Zhiwei Ma; Julie Laux; Rafael Villasmil; Shaheen N. Khan; Sheikh Riazuddin; Javed Akram; Robert N. Cole; C. Conover Talbot; Nader Pourmand; Norann A. Zaghloul; J. Fielding Hejtmancik; S. Amer Riazuddin

FOXE3 is a lens-specific transcription factor that has been associated with anterior segment ocular dysgenesis. To determine the transcriptional target(s) of FOXE3 that are indispensable for the anterior segment development, we examined the transcriptome and the proteome of cells expressing truncated FOXE3 responsible for Peters anomaly identified through linkage-coupled next-generation whole-exome sequencing. We found that DNAJB1, an autophagy-associated protein, was the only candidate exhibiting differential expression in both screens. We confirmed the candidacy of DNAJB1 through chromatin immunoprecipitation and luciferase assays while knockdown of DNAJB1 in human lens epithelial cells resulted in a mitotic arrest. Subsequently, we targeted dnajb1a in zebrafish through injection of a splice-blocking morpholino. The dnajb1a morphants exhibited underdeveloped cataractous lenses with persistent apoptotic nuclei. In conclusion, here we report DNAJB1 is a transcriptional target of FOXE3 in a novel pathway that is crucial for the development of the anterior segment of the eye.


PLOS ONE | 2015

Missense Mutations in CRYAB Are Liable for Recessive Congenital Cataracts.

Xiaodong Jiaox; Shahid Y. Khan; Bushra Irum; Arif O. Khan; Qiwei Wang; Firoz Kabir; Asma A. Khan; Tayyab Husnain; Javed Akram; Sheikh Riazuddin; J. Fielding Hejtmancik; S. Amer Riazuddin

Purpose This study was initiated to identify causal mutations responsible for autosomal recessive congenital cataracts in consanguineous familial cases. Methods Affected individuals underwent a detailed ophthalmological and clinical examination, and slit-lamp photographs were ascertained for affected individuals who have not yet been operated for the removal of the cataractous lens. Blood samples were obtained, and genomic DNA was extracted from white blood cells. A genome-wide scan was completed with short tandem repeat (STR) markers, and the logarithm of odds (LOD) scores were calculated. Protein coding exons of CRYAB were sequenced, bi-directionally. Evolutionary conservation was investigated by aligning CRYAB orthologues, and the expression of Cryab in embryonic and postnatal mice lens was investigated with TaqMan probe. Results The clinical and ophthalmological examinations suggested that all affected individuals had nuclear cataracts. Genome-wide linkage analysis suggested a potential region on chromosome 11q23 harboring CRYAB. DNA sequencing identified a missense variation: c.34C>T (p.R12C) in CRYAB that segregated with the disease phenotype in the family. Subsequent interrogation of our entire cohort of familial cases identified a second familial case localized to chromosome 11q23 harboring a c.31C>T (p.R11C) mutation. In silico analyses suggested that the mutations identified in familial cases, p.R11C and p.R12C will not be tolerated by the three-dimensional structure of CRYAB. Real-time PCR analysis identified the expression of Cryab in mouse lens as early as embryonic day 15 (E15) that increased significantly until postnatal day 6 (P6) with steady level of expression thereafter. Conclusion Here, we report two novel missense mutations, p.R11C and p.R12C, in CRYAB associated with autosomal recessive congenital nuclear cataracts.


PLOS ONE | 2016

Mutation in LIM2 Is Responsible for Autosomal Recessive Congenital Cataracts.

Bushra Irum; Shahid Y. Khan; Muhammad Ali; Haiba Kaul; Firoz Kabir; Bushra Rauf; Fareeha Fatima; Raheela Nadeem; Arif O. Khan; Saif Al Obaisi; Muhammad Asif Naeem; Idrees Ahmad Nasir; Shaheen N. Khan; Tayyab Husnain; Sheikh Riazuddin; Javed Akram; Allen O. Eghrari; S. Amer Riazuddin

Purpose To identify the molecular basis of non-syndromic autosomal recessive congenital cataracts (arCC) in a consanguineous family. Methods All family members participating in the study received a comprehensive ophthalmic examination to determine their ocular phenotype and contributed a blood sample, from which genomic DNA was extracted. Available medical records and interviews with the family were used to compile the medical history of the family. The symptomatic history of the individuals exhibiting cataracts was confirmed by slit-lamp biomicroscopy. A genome-wide linkage analysis was performed to localize the disease interval. The candidate gene, LIM2 (lens intrinsic membrane protein 2), was sequenced bi-directionally to identify the disease-causing mutation. The physical changes caused by the mutation were analyzed in silico through homology modeling, mutation and bioinformatic algorithms, and evolutionary conservation databases. The physiological importance of LIM2 to ocular development was assessed in vivo by real-time expression analysis of Lim2 in a mouse model. Results Ophthalmic examination confirmed the diagnosis of nuclear cataracts in the affected members of the family; the inheritance pattern and cataract development in early infancy indicated arCC. Genome-wide linkage analysis localized the critical interval to chromosome 19q with a two-point logarithm of odds (LOD) score of 3.25. Bidirectional sequencing identified a novel missense mutation, c.233G>A (p.G78D) in LIM2. This mutation segregated with the disease phenotype and was absent in 192 ethnically matched control chromosomes. In silico analysis predicted lower hydropathicity and hydrophobicity but higher polarity of the mutant LIM2-encoded protein (MP19) compared to the wild-type. Moreover, these analyses predicted that the mutation would disrupt the secondary structure of a transmembrane domain of MP19. The expression of Lim2, which was detected in the mouse lens as early as embryonic day 15 (E15) increased after birth to a level that was sustained through the postnatal time points. Conclusion A novel missense mutation in LIM2 is responsible for autosomal recessive congenital cataracts.


PLOS ONE | 2016

A Common Ancestral Mutation in CRYBB3 Identified in Multiple Consanguineous Families with Congenital Cataracts.

Xiaodong Jiao; Firoz Kabir; Bushra Irum; Arif O. Khan; Qiwei Wang; David Li; Asma A. Khan; Tayyab Husnain; Javed Akram; Sheikh Riazuddin; J. Fielding Hejtmancik; S. Amer Riazuddin

Purpose This study was performed to investigate the genetic determinants of autosomal recessive congenital cataracts in large consanguineous families. Methods Affected individuals underwent a detailed ophthalmological examination and slit-lamp photographs of the cataractous lenses were obtained. An aliquot of blood was collected from all participating family members and genomic DNA was extracted from white blood cells. Initially, a genome-wide scan was performed with genomic DNAs of family PKCC025 followed by exclusion analysis of our familial cohort of congenital cataracts. Protein-coding exons of CRYBB1, CRYBB2, CRYBB3, and CRYBA4 were sequenced bidirectionally. A haplotype was constructed with SNPs flanking the causal mutation for affected individuals in all four families, while the probability that the four familial cases have a common founder was estimated using EM and CHM-based algorithms. The expression of Crybb3 in the developing murine lens was investigated using TaqMan assays. Results The clinical and ophthalmological examinations suggested that all affected individuals had nuclear cataracts. Genome-wide linkage analysis localized the causal phenotype in family PKCC025 to chromosome 22q with statistically significant two-point logarithm of odds (LOD) scores. Subsequently, we localized three additional families, PKCC063, PKCC131, and PKCC168 to chromosome 22q. Bidirectional Sanger sequencing identified a missense variation: c.493G>C (p.Gly165Arg) in CRYBB3 that segregated with the disease phenotype in all four familial cases. This variation was not found in ethnically matched control chromosomes, the NHLBI exome variant server, or the 1000 Genomes or dbSNP databases. Interestingly, all four families harbor a unique disease haplotype that strongly suggests a common founder of the causal mutation (p<1.64E-10). We observed expression of Crybb3 in the mouse lens as early as embryonic day 15 (E15), and expression remained relatively steady throughout development. Conclusion Here, we report a common ancestral mutation in CRYBB3 associated with autosomal recessive congenital cataracts identified in four familial cases of Pakistani origin.


Human genome variation | 2016

A spectrum of CYP1B1 mutations associated with primary congenital glaucoma in families of Pakistani descent

Bushra Rauf; Bushra Irum; Firoz Kabir; Sabika Firasat; Muhammad Asif Naeem; Shaheen N. Khan; Tayyab Husnain; Sheikh Riazuddin; Javed Akram; S. Amer Riazuddin

Glaucoma is the second leading cause of blindness, affecting ~65 million people worldwide. We identified and ascertained a large cohort of inbred families with multiple individuals manifesting cardinal symptoms of primary congenital glaucoma (PCG) to investigate the etiology of the disease at a molecular level. Ophthalmic examinations, including slit-lamp microscopy and applanation tonometry, were performed to characterize the causal phenotype and confirm that affected individuals fulfilled the diagnostic criteria for PCG. Subsequently, exclusion analysis was completed with fluorescently labeled short tandem repeat markers, followed by Sanger sequencing to identify pathogenic variants. Exclusion analysis suggested linkage to the CYP1B1 locus, with positive two-point logarithm of odds scores in 23 families, while Sanger sequencing identified a total of 11 variants, including two novel mutations, in 23 families. All mutations segregated with the disease phenotype in their respective families. These included the following seven missense mutations: p.Y81N, p.E229K, p.R368H, p.R390H, p.W434R, p.R444Q and p.R469W, as well as one nonsense mutation, p.Q37*, and three frameshift mutations, p.W246Lfs81*, p.T404Sfs30* and p.P442Qfs15*. In conclusion, we identified a total of 11 mutations, reconfirming the genetic heterogeneity of CYP1B1 in the pathogenesis of PCG. To the best of our knowledge, this is the largest study investigating the contribution of CYP1B1 to the pathogenesis of PCG in the Pakistani population.


PLOS ONE | 2016

Deletion at the GCNT2 Locus Causes Autosomal Recessive Congenital Cataracts

Bushra Irum; Shahid Y. Khan; Muhammad Ali; Muhammad Daud; Firoz Kabir; Bushra Rauf; Fareeha Fatima; Hira Iqbal; Arif O. Khan; Saif Al Obaisi; Muhammad Asif Naeem; Idrees Ahmad Nasir; Shaheen N. Khan; Tayyab Husnain; Sheikh Riazuddin; Javed Akram; Allen O. Eghrari; S. Amer Riazuddin

Purpose The aim of this study is to identify the molecular basis of autosomal recessive congenital cataracts (arCC) in a large consanguineous pedigree. Methods All participating individuals underwent a detailed ophthalmic examination. Each patient’s medical history, particularly of cataracts and other ocular abnormalities, was compiled from available medical records and interviews with family elders. Blood samples were donated by all participating family members and used to extract genomic DNA. Genetic analysis was performed to rule out linkage to known arCC loci and genes. Whole-exome sequencing libraries were prepared and paired-end sequenced. A large deletion was found that segregated with arCC in the family, and chromosome walking was conducted to estimate the proximal and distal boundaries of the deletion mutation. Results Exclusion and linkage analysis suggested linkage to a region of chromosome 6p24 harboring GCNT2 (glucosaminyl (N-acetyl) transferase 2) with a two-point logarithm of odds score of 5.78. PCR amplifications of the coding exons of GCNT2 failed in individuals with arCC, and whole-exome data analysis revealed a large deletion on chromosome 6p in the region harboring GCNT2. Chromosomal walking using multiple primer pairs delineated the extent of the deletion to approximately 190 kb. Interestingly, a failure to amplify a junctional fragment of the deletion break strongly suggests an insertion in addition to the large deletion. Conclusion Here, we report a novel insertion/deletion mutation at the GCNT2 locus that is responsible for congenital cataracts in a large consanguineous family.


Investigative Ophthalmology & Visual Science | 2018

Proteome Profiling of Developing Murine Lens Through Mass Spectrometry

Shahid Y. Khan; Muhammad Ali; Firoz Kabir; Santosh Renuse; Chan Hyun Na; C. Conover Talbot; Sean F. Hackett; S. Amer Riazuddin

Purpose We previously completed a comprehensive profile of the mouse lens transcriptome. Here, we investigate the proteome of the mouse lens through mass spectrometry–based protein sequencing at the same embryonic and postnatal time points. Methods We extracted mouse lenses at embryonic day 15 (E15) and 18 (E18) and postnatal day 0 (P0), 3 (P3), 6 (P6), and 9 (P9). The lenses from each time point were preserved in three distinct pools to serve as biological replicates for each developmental stage. The total cellular protein was extracted from the lens, digested with trypsin, and labeled with isobaric tandem mass tags (TMT) for three independent TMT experiments. Results A total of 5404 proteins were identified in the mouse ocular lens in at least one TMT set, 4244 in two, and 3155 were present in all three TMT sets. The majority of the proteins exhibited steady expression at all six developmental time points; nevertheless, we identified 39 proteins that exhibited an 8-fold differential (higher or lower) expression during the developmental time course compared to their respective levels at E15. The lens proteome is composed of diverse proteins that have distinct biological properties and functional characteristics, including proteins associated with cataractogenesis and autophagy. Conclusions We have established a comprehensive profile of the developing murine lens proteome. This repository will be helpful in identifying critical components of lens development and processes essential for the maintenance of its transparency.


Scientific Data | 2018

Whole genome sequencing data for two individuals of Pakistani descent

Shahid Y. Khan; Firoz Kabir; Oussama M’Hamdi; Xiaodong Jiao; Muhammad Asif Naeem; Shaheen N. Khan; Sheikh Riazuddin; J. Fielding Hejtmancik; S. Amer Riazuddin

Here we report next-generation based whole genome sequencing of two individuals (H1 and H2) from a family of Pakistani descent. The genomic DNA was used to prepare paired-end libraries for whole-genome sequencing. Deep sequencing yielded 706.49 and 778.12 million mapped reads corresponding to 70.64 and 77.81 Gb sequence data and 23× and 25× average coverage for H1 and H2, respectively. Notably, a total of 448,544 and 470,683 novel variants, not present in the single nucleotide polymorphism database (dbSNP), were identified in H1 and H2, respectively. Comparative analysis identified 2,415,852 variants common in both genomes including 240,181 variants absent in the dbSNP. Principal component analysis linked the ancestry of both genomes with South Asian populations. In conclusion, we report whole genome sequences of two individuals from a family of Pakistani descent.


PLOS Genetics | 2018

Mutation in the intracellular chloride channel CLCC1 associated with autosomal recessive retinitis pigmentosa

Lin Li; Xiaodong Jiao; Ilaria D’Atri; Fumihito Ono; Ralph Nelson; Chi-Chao Chan; Naoki Nakaya; Zhiwei Ma; Yan Ma; Xiaoying Cai; Longhua Zhang; Siying Lin; Abdul Hameed; Barry A. Chioza; Holly Hardy; Gavin Arno; Sarah Hull; Muhammad Imran Khan; James Fasham; Gaurav V. Harlalka; Michel Michaelides; Anthony T. Moore; Zeynep Coban Akdemir; Shalini N. Jhangiani; James R. Lupski; Frans P.M. Cremers; Raheel Qamar; Ahmed Salman; John K. Chilton; Jay E. Self

We identified a homozygous missense alteration (c.75C>A, p.D25E) in CLCC1, encoding a presumptive intracellular chloride channel highly expressed in the retina, associated with autosomal recessive retinitis pigmentosa (arRP) in eight consanguineous families of Pakistani descent. The p.D25E alteration decreased CLCC1 channel function accompanied by accumulation of mutant protein in granules within the ER lumen, while siRNA knockdown of CLCC1 mRNA induced apoptosis in cultured ARPE-19 cells. TALEN KO in zebrafish was lethal 11 days post fertilization. The depressed electroretinogram (ERG) cone response and cone spectral sensitivity of 5 dpf KO zebrafish and reduced eye size, retinal thickness, and expression of rod and cone opsins could be rescued by injection of wild type CLCC1 mRNA. Clcc1+/- KO mice showed decreased ERGs and photoreceptor number. Together these results strongly suggest that intracellular chloride transport by CLCC1 is a critical process in maintaining retinal integrity, and CLCC1 is crucial for survival and function of retinal cells.


Experimental Eye Research | 2018

Comparative transcriptome analysis of hESC- and iPSC-derived corneal endothelial cells

Muhammad Ali; Shahid Y. Khan; Firoz Kabir; John D. Gottsch; S. Amer Riazuddin

&NA; The corneal endothelium (CE), a monolayer of hexagonal cells constitutes the innermost layer of the cornea that is critical in maintaining clarity by mediating hydration through barrier and pump functions. Corneal endothelial cells (CECs) have limited proliferative potential and therefore generation of CECs has been undertaken by many groups. We previously reported generation of CECs from peripheral blood mononuclear cell (PBMC)‐originated, induced pluripotent stem cells (iPSCs). In here, we extend our analysis through next‐generation seqeuncing based transcriptome profiling of H9 human embryonic stem cell (hESC)‐ and human PBMC‐originated, iPSC‐derived CECs. The differentiating CECs on day 20 (D20) exhibited a tightly packed hexagonal/polygonal shape expressing zona occludens‐1 (ZO‐1) and N‐cadherin at the cell boundaries. Next‐generation RNA sequencing of hESC‐ and iPSC‐derived CECs detected expression (≥0.659 RPKM) of 13,546 and 13,536 genes, respectively. Comparative transcriptome analysis of hESC‐ and iPSC‐derived CECs revealed 13,208 (>96%) genes common in both transcriptomes. Among the 13,208 genes common in these transcriptomes, 12,580 (>95%) exhibited a quantitatively similar expression. To the best of our knowledge, this is the first report presenting comparative transcriptome analysis of hESC‐ and iPSC‐derived CECs. HighlightWe previously reported generation of corenal endothelial cells (CECs) fromperipheral blood mononuclear cell (PBMC)‐originated, induced pluripotent stem cells (iPSCs).In here, we extend our analysis through next‐generation seqeuncing based transcriptome profiling of H9 human embryonic stem cells (hESC)‐ and human PBMC‐originated, iPSC‐derived CECs.Comparative transcriptome analysis of hESC‐ and iPSC‐derived CECs identified >96% genes common in both transcriptomes.

Collaboration


Dive into the Firoz Kabir's collaboration.

Top Co-Authors

Avatar

S. Amer Riazuddin

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shahid Y. Khan

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Javed Akram

University of Health Sciences Lahore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tayyab Husnain

University of the Punjab

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaodong Jiao

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge