Network


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

Hotspot


Dive into the research topics where Paul Ling-Fung Tang is active.

Publication


Featured researches published by Paul Ling-Fung Tang.


Clinical Genetics | 2015

Exome Sequencing in 32 Patients with Anophthalmia/Microphthalmia and Developmental Eye Defects

Anne Slavotinek; S.T. Garcia; G. Chandratillake; Tanya Bardakjian; E. Ullah; Di Wu; K. Umeda; Richard Lao; Paul Ling-Fung Tang; Eunice Wan; Lohith Madireddy; S. Lyalina; Bryce A. Mendelsohn; S. Dugan; J. Tirch; R. Tischler; J. Harris; M.J. Clark; S. Chervitz; A. Patwardhan; J.M. West; Philip C. Ursell; A. de Alba Campomanes; Adele Schneider; Pui-Yan Kwok; Sergio E. Baranzini; R.O. Chen

Anophthalmia/microphthalmia (A/M) is a genetically heterogeneous birth defect for which the etiology is unknown in more than 50% of patients. We used exome sequencing with the ACE ExomeTM (Personalis, Inc; 18 cases) and UCSF Genomics Core (21 cases) to sequence 28 patients with A/M and four patients with varied developmental eye defects. In the 28 patients with A/M, we identified de novo mutations in three patients (OTX2, p.(Gln91His), RARB, p.Arg387Cys and GDF6, p.Ala249Glu) and inherited mutations in STRA6 in two patients. In patients with developmental eye defects, a female with cataracts and cardiomyopathy had a de novo COL4A1 mutation, p.(Gly773Arg), expanding the phenotype associated with COL4A1 to include cardiomyopathy. A male with a chorioretinal defect, microcephaly, seizures and sensorineural deafness had two PNPT1 mutations, p.(Ala507Ser) and c.401‐1G>A, and we describe eye defects associated with this gene for the first time. Exome sequencing was efficient for identifying mutations in pathogenic genes for which there is no clinical testing available and for identifying cases that expand phenotypic spectra, such as the PNPT1 and COL4A1‐associated disorders described here.


Human Molecular Genetics | 2013

Focal facial dermal dysplasia, type IV, is caused by mutations in CYP26C1

Anne Slavotinek; Pavni Mehrotra; Irina Nazarenko; Paul Ling-Fung Tang; Richard Lao; Don Cameron; Ben Li; Catherine Chu; Chris Chou; Ann L Marqueling; Mani Yahyavi; Kelly M. Cordoro; Ilona J. Frieden; Tom Glaser; Trine Prescott; Marie-Anne Morren; Koenraad Devriendt; Pui-Yan Kwok; Martin Petkovich; Robert J. Desnick

Focal facial dermal dysplasia (FFDD) Type IV is a rare syndrome characterized by facial lesions resembling aplasia cutis in a preauricular distribution along the line of fusion of the maxillary and mandibular prominences. To identify the causative gene(s), exome sequencing was performed in a family with two affected siblings. Assuming autosomal recessive inheritance, two novel sequence variants were identified in both siblings in CYP26C1-a duplication of seven base pairs, which was maternally inherited, c.844_851dupCCATGCA, predicting p.Glu284fsX128 and a missense mutation, c.1433G>A, predicting p.Arg478His, that was paternally inherited. The duplication predicted a frameshift mutation that led to a premature stop codon and premature chain termination, whereas the missense mutation was not functional based on its in vitro expression in mammalian cells. The FFDD skin lesions arise along the sites of fusion of the maxillary and mandibular prominences early in facial development, and Cyp26c1 was expressed exactly along the fusion line for these facial prominences in the first branchial arch in mice. Sequencing of four additional, unrelated Type IV FFDD patients and eight Type II or III TWIST2-negative FFDD patients revealed that three of the Type IV patients were homozygous for the duplication, whereas none of the Type II or III patients had CYP26C1 mutations. The seven base pairs duplication was present in 0.3% of healthy controls and 0.3% of patients with other birth defects. These findings suggest that the phenotypic manifestations of FFDD Type IV can be non-penetrant or underascertained. Thus, FFDD Type IV results from the loss of function mutations in CYP26C1.


Journal of Bone and Mineral Research | 2009

Genome‐Wide Haplotype Association Mapping in Mice Identifies a Genetic Variant in CER1 Associated With BMD and Fracture in Southern Chinese Women

Paul Ling-Fung Tang; Ching-Lung Cheung; Pak Sham; Philip McClurg; Bob Lee; Shut-Yan Chan; David K. Smith; Julian A. Tanner; Andrew I. Su; Kathryn S. E. Cheah; Annie W. C. Kung; You-Qiang Song

BMD is a heritable trait and risk indicator for osteoporosis. In this study, we used a genome‐wide haplotype association mapping (HAM) approach to identify a haplotype block within Cer1 that partitions inbred mice strains into high and low BMD groups. A cohort of 1083 high and low BMD human subjects were studied, and a nonsynonymous SNP (rs3747532) in human CER1 was identified to be associated with increased risk of both low BMD in premenopausal women (OR: 2.2; 95% CI: 1.0–4.6; p < 0.05) and increased risk of vertebral fractures (OR: 1.82, p = 0.025) in the postmenopausal cohort. We also showed that Cer1 is expressed in mouse bone and growth plate by RT‐PCR, immunohistochemistry, and in situ hybridization, consistent with polymorphisms potentially influencing BMD. Our successful identification of an association with CER1 in humans together with our mouse study suggests that CER1 may play a role in the development of bone or its metabolism. Our study highlights the use of publicly available databases for rapidly surveying the genome for quantitative trait loci.


Genome Biology | 2014

Prioritizing causal disease genes using unbiased genomic features

Rahul C. Deo; Gabriel Musso; Murat Tasan; Paul Ling-Fung Tang; Annie Poon; Christiana Yuan; Janine F. Felix; Rameen Beroukhim; Teresa De Marco; Pui-Yan Kwok; Calum A. MacRae; Frederick P. Roth

BackgroundCardiovascular disease (CVD) is the leading cause of death in the developed world. Human genetic studies, including genome-wide sequencing and SNP-array approaches, promise to reveal disease genes and mechanisms representing new therapeutic targets. In practice, however, identification of the actual genes contributing to disease pathogenesis has lagged behind identification of associated loci, thus limiting the clinical benefits.ResultsTo aid in localizing causal genes, we develop a machine learning approach, Objective Prioritization for Enhanced Novelty (OPEN), which quantitatively prioritizes gene-disease associations based on a diverse group of genomic features. This approach uses only unbiased predictive features and thus is not hampered by a preference towards previously well-characterized genes. We demonstrate success in identifying genetic determinants for CVD-related traits, including cholesterol levels, blood pressure, and conduction system and cardiomyopathy phenotypes. Using OPEN, we prioritize genes, including FLNC, for association with increased left ventricular diameter, which is a defining feature of a prevalent cardiovascular disorder, dilated cardiomyopathy or DCM. Using a zebrafish model, we experimentally validate FLNC and identify a novel FLNC splice-site mutation in a patient with severe DCM.ConclusionOur approach stands to assist interpretation of large-scale genetic studies without compromising their fundamentally unbiased nature.


Neuron | 2015

Pbx Regulates Patterning of the Cerebral Cortex in Progenitors and Postmitotic Neurons

Olga Golonzhka; Alex S. Nord; Paul Ling-Fung Tang; Susan Lindtner; Athena R. Ypsilanti; Elisabetta Ferretti; Axel Visel; Licia Selleri; John L.R. Rubenstein

We demonstrate using conditional mutagenesis that Pbx1, with and without Pbx2(+/-) sensitization, regulates regional identity and laminar patterning of the developing mouse neocortex in cortical progenitors (Emx1-Cre) and in newly generated neurons (Nex1-Cre). Pbx1/2 mutants have three salient molecular phenotypes of cortical regional and laminar organization: hypoplasia of the frontal cortex, ventral expansion of the dorsomedial cortex, and ventral expansion of Reelin expression in the cortical plate of the frontal cortex, concomitant with an inversion of cortical layering in the rostral cortex. Molecular analyses, including PBX ChIP-seq, provide evidence that PBX promotes frontal cortex identity by repressing genes that promote dorsocaudal fate.


European Journal of Human Genetics | 2015

Novel mutations in PXDN cause microphthalmia and anterior segment dysgenesis

Alex Choi; Richard Lao; Paul Ling-Fung Tang; Eunice Wan; Wasima Mayer; Tanya Bardakjian; Gary M. Shaw; Pui-Yan Kwok; Adele Schneider; Anne Slavotinek

We used exome sequencing to study a non-consanguineous family with two children who had anterior segment dysgenesis, sclerocornea, microphthalmia, hypotonia and developmental delays. Sanger sequencing verified two Peroxidasin (PXDN) mutations in both sibs—a maternally inherited, nonsense mutation, c.1021C>T predicting p.(Arg341*), and a paternally inherited, 23-basepair deletion causing a frameshift and premature protein truncation, c.2375_2397del23, predicting p.(Leu792Hisfs*67). We re-examined exome data from 20 other patients with structural eye defects and identified two additional PXDN mutations in a sporadic male with bilateral microphthalmia, cataracts and anterior segment dysgenesis—a maternally inherited, frameshift mutation, c.1192delT, predicting p.(Tyr398Thrfs*40) and a paternally inherited, missense substitution that was predicted to be deleterious, c.947 A>C, predicting p.(Gln316Pro). Mutations in PXDN were previously reported in three families with congenital cataracts, microcornea, sclerocornea and developmental glaucoma. The gene is expressed in corneal epithelium and is secreted into the extracellular matrix. Defective peroxidasin has been shown to impair sulfilimine bond formation in collagen IV, a constituent of the basement membrane, implying that the eye defects result because of loss of basement membrane integrity in the developing eye. Our finding of a broader phenotype than previously appreciated for PXDN mutations is typical for exome-sequencing studies, which have proven to be highly effective for mutation detection in patients with atypical presentations. We conclude that PXDN sequencing should be considered in microphthalmia with anterior segment dysgenesis.


Human Molecular Genetics | 2015

DLX4 is associated with orofacial clefting and abnormal jaw development

Di Wu; Shyamali Mandal; Alex Choi; August Anderson; Michaela Prochazkova; Hazel Perry; Vera Lúcia Gil-da-Silva-Lopes; Richard Lao; Eunice Wan; Paul Ling-Fung Tang; Pui-Yan Kwok; Ophir D. Klein; Bian Zhuan; Anne Slavotinek

Cleft lip and/or palate (CL/P) are common structural birth defects in humans. We used exome sequencing to study a patient with bilateral CL/P and identified a single nucleotide deletion in the patient and her similarly affected son—c.546_546delG, predicting p.Gln183Argfs*57 in the Distal-less 4 (DLX4) gene. The sequence variant was absent from databases, predicted to be deleterious and was verified by Sanger sequencing. In mammals, there are three Dlx homeobox clusters with closely located gene pairs (Dlx1/Dlx2, Dlx3/Dlx4, Dlx5/Dlx6). In situ hybridization showed that Dlx4 was expressed in the mesenchyme of the murine palatal shelves at E12.5, prior to palate closure. Wild-type human DLX4, but not mutant DLX4_c.546delG, could activate two murine Dlx conserved regulatory elements, implying that the mutation caused haploinsufficiency. We showed that reduced DLX4 expression after short interfering RNA treatment in a human cell line resulted in significant up-regulation of DLX3, DLX5 and DLX6, with reduced expression of DLX2 and significant up-regulation of BMP4, although the increased BMP4 expression was demonstrated only in HeLa cells. We used antisense morpholino oligonucleotides to target the orthologous Danio rerio gene, dlx4b, and found reduced cranial size and abnormal cartilaginous elements. We sequenced DLX4 in 155 patients with non-syndromic CL/P and CP, but observed no sequence variants. From the published literature, Dlx1/Dlx2 double homozygous null mice and Dlx5 homozygous null mice both have clefts of the secondary palate. This first finding of a DLX4 mutation in a family with CL/P establishes DLX4 as a potential cause of human clefts.


Development | 2015

Endothelin signaling activates Mef2c expression in the neural crest through a MEF2C-dependent positive-feedback transcriptional pathway

Jianxin Hu; Michael P. Verzi; Ashley S. Robinson; Paul Ling-Fung Tang; Lisa L. Hua; Shan-Mei Xu; Pui-Yan Kwok; Brian L. Black

Endothelin signaling is essential for neural crest development, and dysregulated Endothelin signaling is associated with several neural crest-related disorders, including Waardenburg and other syndromes. However, despite the crucial roles of this pathway in neural crest development and disease, the transcriptional effectors directly activated by Endothelin signaling during neural crest development remain incompletely elucidated. Here, we establish that the MADS box transcription factor MEF2C is an immediate downstream transcriptional target and effector of Endothelin signaling in the neural crest. We show that Endothelin signaling activates Mef2c expression in the neural crest through a conserved enhancer in the Mef2c locus and that CRISPR-mediated deletion of this Mef2c neural crest enhancer from the mouse genome abolishes Endothelin induction of Mef2c expression. Moreover, we demonstrate that Endothelin signaling activates neural crest expression of Mef2c by de-repressing MEF2C activity through a Calmodulin-CamKII-histone deacetylase signaling cascade. Thus, these findings identify a MEF2C-dependent, positive-feedback mechanism for Endothelin induction and establish MEF2C as an immediate transcriptional effector and target of Endothelin signaling in the neural crest. Summary: The MADS box transcription factor MEF2C is identified as an immediate downstream target of endothelin and is part of a positive-feedback mechanism for endothelin induction in the neural crest.


Arthritis & Rheumatism | 2016

Brief Report: Whole-Exome Sequencing for Identification of Potential Causal Variants for Diffuse Cutaneous Systemic Sclerosis.

Angel C. Y. Mak; Paul Ling-Fung Tang; Clare M. Cleveland; Melanie H. Smith; M. Kari Connolly; Tamiko R. Katsumoto; Paul J. Wolters; Pui-Yan Kwok; Lindsey A. Criswell

Scleroderma is a genetically complex autoimmune disease with substantial phenotypic heterogeneity. Previous genome‐wide association studies have identified common genetic variants associated with disease risk, but these studies are not designed to capture rare or potential causal variants. Our goal was to identify rare as well as common genetic variants in patients with diffuse cutaneous systemic sclerosis (dcSSc) through whole‐exome sequencing (WES) in order to identify potential causal variants.


JCI insight | 2018

Mutations in Hnrnpa1 cause congenital heart defects

Zhe Yu; Paul Ling-Fung Tang; Jing Wang; Suying Bao; Joseph T.C. Shieh; Alan W.L. Leung; Zhao Zhang; Fei Gao; Sandra Y.Y. Wong; Andy L.C. Hui; Yuan Gao; Nelson W. F. Dung; Zhi-Gang Zhang; Yanhui Fan; Xueya Zhou; Yalun Zhang; Dana S.M. Wong; Pc Sham; Abid Azhar; Pui-Yan Kwok; Patrick P.L. Tam; Qizhou Lian; Kathryn S. E. Cheah; Binbin Wang; You-Qiang Song

Incomplete penetrance of congenital heart defects (CHDs) was observed in a mouse model. We hypothesized that the contribution of a major genetic locus modulates the manifestation of the CHDs. After genome-wide linkage mapping, fine mapping, and high-throughput targeted sequencing, a recessive frameshift mutation of the heterogeneous nuclear ribonucleoprotein A1 (Hnrnpa1) gene was confirmed (Hnrnpa1ct). Hnrnpa1 was expressed in both the first heart field (FHF) and second heart field (SHF) at the cardiac crescent stage but was only maintained in SHF progenitors after heart tube formation. Hnrnpa1ct/ct homozygous mutants displayed complete CHD penetrance, including truncated and incomplete looped heart tube at E9.5, ventricular septal defect (VSD) and persistent truncus arteriosus (PTA) at E13.5, and VSD and double outlet right ventricle at P0. Impaired development of the dorsal mesocardium and sinoatrial node progenitors was also observed. Loss of Hnrnpa1 expression leads to dysregulation of cardiac transcription networks and multiple signaling pathways, including BMP, FGF, and Notch in the SHF. Finally, two rare heterozygous mutations of HNRNPA1 were detected in human CHDs. These findings suggest a role of Hnrnpa1 in embryonic heart development in mice and humans.

Collaboration


Dive into the Paul Ling-Fung Tang's collaboration.

Top Co-Authors

Avatar

Pui-Yan Kwok

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard Lao

University of California

View shared research outputs
Top Co-Authors

Avatar

Eunice Wan

University of California

View shared research outputs
Top Co-Authors

Avatar

Adele Schneider

Albert Einstein Medical Center

View shared research outputs
Top Co-Authors

Avatar

Di Wu

University of California

View shared research outputs
Top Co-Authors

Avatar

Tanya Bardakjian

Albert Einstein Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alex Choi

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge