Network


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

Hotspot


Dive into the research topics where Huanan Ren is active.

Publication


Featured researches published by Huanan Ren.


Nature Genetics | 2012

Mutations in NMNAT1 cause Leber congenital amaurosis and identify a new disease pathway for retinal degeneration

Robert K. Koenekoop; Hui Wang; Jacek Majewski; Xia Wang; Irma Lopez; Huanan Ren; Yiyun Chen; Yumei Li; Gerald A. Fishman; Mohammed Genead; Jeremy Schwartzentruber; Naimesh Solanki; Elias I. Traboulsi; Jingliang Cheng; Clare V. Logan; Martin McKibbin; Bruce E. Hayward; David A. Parry; Colin A. Johnson; Mohammed Nageeb; James A. Poulter; Moin D. Mohamed; Hussain Jafri; Yasmin Rashid; Graham R. Taylor; Vafa Keser; Graeme Mardon; Huidan Xu; Chris F. Inglehearn; Qing Fu

Leber congenital amaurosis (LCA) is a blinding retinal disease that presents within the first year after birth. Using exome sequencing, we identified mutations in the nicotinamide adenine dinucleotide (NAD) synthase gene NMNAT1 encoding nicotinamide mononucleotide adenylyltransferase 1 in eight families with LCA, including the family in which LCA was originally linked to the LCA9 locus. Notably, all individuals with NMNAT1 mutations also have macular colobomas, which are severe degenerative entities of the central retina (fovea) devoid of tissue and photoreceptors. Functional assays of the proteins encoded by the mutant alleles identified in our study showed that the mutations reduce the enzymatic activity of NMNAT1 in NAD biosynthesis and affect protein folding. Of note, recent characterization of the slow Wallerian degeneration (Wlds) mouse model, in which prolonged axonal survival after injury is observed, identified NMNAT1 as a neuroprotective protein when ectopically expressed. Our findings identify a new disease mechanism underlying LCA and provide the first link between endogenous NMNAT1 dysfunction and a human nervous system disorder.


Human Genetics | 2014

Next generation sequencing-based molecular diagnosis of retinitis pigmentosa: identification of a novel genotype-phenotype correlation and clinical refinements

Feng Wang; Hui Wang; Han Fang Tuan; Duy Nguyen; Vincent Sun; Vafa Keser; Sara J. Bowne; Lori S. Sullivan; Hongrong Luo; Ling Zhao; Xia Wang; Jacques Zaneveld; Jason S. Salvo; Sorath Noorani Siddiqui; Louise Mao; Dianna K. Wheaton; David G. Birch; Kari Branham; John R. Heckenlively; Cindy Wen; Ken Flagg; Henry A. Ferreyra; Jacqueline Pei; Ayesha Khan; Huanan Ren; Keqing Wang; Irma Lopez; Raheel Qamar; Juan Carlos Zenteno; Raul Ayala-Ramirez

Abstract Retinitis pigmentosa (RP) is a devastating form of retinal degeneration, with significant social and professional consequences. Molecular genetic information is invaluable for an accurate clinical diagnosis of RP due to its high genetic and clinical heterogeneity. Using a gene capture panel that covers 163 of the currently known retinal disease genes, including 48 RP genes, we performed a comprehensive molecular screening in a collection of 123 RP unsettled probands from a wide variety of ethnic backgrounds, including 113 unrelated simplex and 10 autosomal recessive RP (arRP) cases. As a result, 61 mutations were identified in 45 probands, including 38 novel pathogenic alleles. Interestingly, we observed that phenotype and genotype were not in full agreement in 21 probands. Among them, eight probands were clinically reassessed, resulting in refinement of clinical diagnoses for six of these patients. Finally, recessive mutations in CLN3 were identified in five retinal degeneration patients, including four RP probands and one cone-rod dystrophy patient, suggesting that CLN3 is a novel non-syndromic retinal disease gene. Collectively, our results underscore that, due to the high molecular and clinical heterogeneity of RP, comprehensive screening of all retinal disease genes is effective in identifying novel pathogenic mutations and provides an opportunity to discover new genotype-phenotype correlations. Information gained from this genetic screening will directly aid in patient diagnosis, prognosis, and treatment, as well as allowing appropriate family planning and counseling.


Journal of Medical Genetics | 2013

Comprehensive molecular diagnosis of 179 Leber congenital amaurosis and juvenile retinitis pigmentosa patients by targeted next generation sequencing.

Xia Wang; Hui Wang; Vincent Sun; Han Fang Tuan; Vafa Keser; Keqing Wang; Huanan Ren; Irma Lopez; Jacques Zaneveld; Sorath Noorani Siddiqui; Stephanie Bowles; Ayesha Khan; Jason S. Salvo; Samuel G. Jacobson; Alessandro Iannaccone; Feng Wang; David G. Birch; John R. Heckenlively; Gerald A. Fishman; Elias I. Traboulsi; Yumei Li; Dianna H. Wheaton; Robert K. Koenekoop; Rui Chen

Background Leber congenital amaurosis (LCA) and juvenile retinitis pigmentosa (RP) are inherited retinal diseases that cause early onset severe visual impairment. An accurate molecular diagnosis can refine the clinical diagnosis and allow gene specific treatments. Methods We developed a capture panel that enriches the exonic DNA of 163 known retinal disease genes. Using this panel, we performed targeted next generation sequencing (NGS) for a large cohort of 179 unrelated and prescreened patients with the clinical diagnosis of LCA or juvenile RP. Systematic NGS data analysis, Sanger sequencing validation, and segregation analysis were utilised to identify the pathogenic mutations. Patients were revisited to examine the potential phenotypic ambiguity at the time of initial diagnosis. Results Pathogenic mutations for 72 patients (40%) were identified, including 45 novel mutations. Of these 72 patients, 58 carried mutations in known LCA or juvenile RP genes and exhibited corresponding phenotypes, while 14 carried mutations in retinal disease genes that were not consistent with their initial clinical diagnosis. We revisited patients in the latter case and found that homozygous mutations in PRPH2 can cause LCA/juvenile RP. Guided by the molecular diagnosis, we reclassified the clinical diagnosis in two patients. Conclusions We have identified a novel gene and a large number of novel mutations that are associated with LCA/juvenile RP. Our results highlight the importance of molecular diagnosis as an integral part of clinical diagnosis.


Nature Genetics | 2015

Mutations in the unfolded protein response regulator ATF6 cause the cone dysfunction disorder achromatopsia

Susanne Kohl; Ditta Zobor; Wei-Chieh Chiang; Nicole Weisschuh; Jennifer Staller; Irene Gonzalez Menendez; Stanley Chang; Susanne C. Beck; Marina Garcia Garrido; Vithiyanjali Sothilingam; Mathias W. Seeliger; Franco Stanzial; Francesco Benedicenti; Francesca Inzana; Elise Héon; Ajoy Vincent; Jill Beis; Tim M. Strom; Günther Rudolph; Susanne Roosing; Anneke I. den Hollander; Frans P.M. Cremers; Irma Lopez; Huanan Ren; Anthony T. Moore; Andrew R. Webster; Michel Michaelides; Robert K. Koenekoop; Eberhart Zrenner; Randal J. Kaufman

Achromatopsia (ACHM) is an autosomal recessive disorder characterized by color blindness, photophobia, nystagmus and severely reduced visual acuity. Using homozygosity mapping and whole-exome and candidate gene sequencing, we identified ten families carrying six homozygous and two compound-heterozygous mutations in the ATF6 gene (encoding activating transcription factor 6A), a key regulator of the unfolded protein response (UPR) and cellular endoplasmic reticulum (ER) homeostasis. Patients had evidence of foveal hypoplasia and disruption of the cone photoreceptor layer. The ACHM-associated ATF6 mutations attenuate ATF6 transcriptional activity in response to ER stress. Atf6−/− mice have normal retinal morphology and function at a young age but develop rod and cone dysfunction with increasing age. This new ACHM-related gene suggests a crucial and unexpected role for ATF6A in human foveal development and cone function and adds to the list of genes that, despite ubiquitous expression, when mutated can result in an isolated retinal photoreceptor phenotype.


American Journal of Human Genetics | 2012

GPR179 is required for depolarizing bipolar cell function and is mutated in autosomal-recessive complete congenital stationary night blindness

Neal S. Peachey; Thomas A. Ray; Ralph J. Florijn; Lucy B. Rowe; Trijntje Sjoerdsma; Susana Contreras-Alcantara; Kenkichi Baba; Gianluca Tosini; Nikita Pozdeyev; P. Michael Iuvone; Pasano Bojang; Jillian N. Pearring; Huibert J. Simonsz; Maria M. van Genderen; David G. Birch; Elias I. Traboulsi; Allison Dorfman; Irma Lopez; Huanan Ren; Andrew F. X. Goldberg; Patsy M. Nishina; Pierre Lachapelle; Maureen A. McCall; Robert K. Koenekoop; Arthur A. B. Bergen; Maarten Kamermans; Ronald G. Gregg

Complete congenital stationary night blindness (cCSNB) is a clinically and genetically heterogeneous group of retinal disorders characterized by nonprogressive impairment of night vision, absence of the electroretinogram (ERG) b-wave, and variable degrees of involvement of other visual functions. We report here that mutations in GPR179, encoding an orphan G protein receptor, underlie a form of autosomal-recessive cCSNB. The Gpr179(nob5/nob5) mouse model was initially discovered by the absence of the ERG b-wave, a component that reflects depolarizing bipolar cell (DBC) function. We performed genetic mapping, followed by next-generation sequencing of the critical region and detected a large transposon-like DNA insertion in Gpr179. The involvement of GPR179 in DBC function was confirmed in zebrafish and humans. Functional knockdown of gpr179 in zebrafish led to a marked reduction in the amplitude of the ERG b-wave. Candidate gene analysis of GPR179 in DNA extracted from patients with cCSNB identified GPR179-inactivating mutations in two patients. We developed an antibody against mouse GPR179, which robustly labeled DBC dendritic terminals in wild-type mice. This labeling colocalized with the expression of GRM6 and was absent in Gpr179(nob5/nob5) mutant mice. Our results demonstrate that GPR179 plays a critical role in DBC signal transduction and expands our understanding of the mechanisms that mediate normal rod vision.


Investigative Ophthalmology & Visual Science | 2013

Next-Generation Sequencing–Based Molecular Diagnosis of a Chinese Patient Cohort With Autosomal Recessive Retinitis Pigmentosa

Qing Fu; Feng Wang; Hui Wang; Fei Xu; Jacques Zaneveld; Huanan Ren; Vafa Keser; Irma Lopez; Han-Fang Tuan; Jason S. Salvo; Xia Wang; Li Zhao; Keqing Wang; Yumei Li; Robert K. Koenekoop; Rui Chen; Ruifang Sui

PURPOSE Retinitis pigmentosa (RP) is a highly heterogeneous genetic disease; therefore, an accurate molecular diagnosis is essential for appropriate disease treatment and family planning. The prevalence of RP in China had been reported at 1 in 3800, resulting in an estimated total of 340,000 Chinese RP patients. However, genetic studies of Chinese RP patients have been very limited. To date, no comprehensive molecular diagnosis has been done for Chinese RP patients. With the emergence of next-generation sequencing (NGS), comprehensive molecular diagnosis of RP is now within reach. The purpose of this study was to perform the first NGS-based comprehensive molecular diagnosis for Chinese RP patients. METHODS Thirty-one well-characterized autosomal recessive RP (arRP) families were recruited. For each family, the DNA sample from one affected member was sequenced using our custom capture panel, which includes 163 retinal disease genes. Variants were called, filtered, and annotated by our in-house automatic pipeline. RESULTS Twelve arRP families were successfully molecular diagnosed, achieving a diagnostic rate of approximately 40%. Interestingly, approximately 63% of the pathogenic mutations we identified are novel, which is higher than that observed in a similar study on European descent (45%). Moreover, the clinical diagnoses of two families were refined based on the pathogenic mutations identified in the patients. CONCLUSIONS We conclude that comprehensive molecular diagnosis can be vital for an accurate clinical diagnosis of RP. Applying this tool on patients from different ethnic groups is essential for enhancing our knowledge of the global spectrum of RP disease-causing mutations.


Journal of Medical Genetics | 2011

A new ocular phenotype associated with an unexpected but known systemic disorder and mutation: novel use of genomic diagnostics and exome sequencing

Jacek Majewski; Zibo Wang; Irma Lopez; Sulaiman Al Humaid; Huanan Ren; Julie Racine; Alex Bazinet; Grant Mitchel; Nancy Braverman; Robert K. Koenekoop

We identified a 28-year-old cognitively normal patient with severe visual loss, absent electrical signals from the photoreceptors by electroretinogram (ERG) and nystagmus due to Leber congenital amaurosis (LCA), associated with hearing loss and Arnold–Chiari malformation. Exome sequencing detected a homozygous PEX1 mutation (p.Gly843Asp). Within a large LCA cohort, we found the mutation again in a 9-month-old baby. Peroxisome biochemical studies on both patients confirmed a peroxisome biogenesis disorder (PBD) in the Zellweger spectrum. We thus demonstrate that these patients, who had isolated LCA on presentation, actually had PBD as the cause of their LCA. Furthermore, the phenotype of the first patient was outside that of a typical Zellweger spectrum, and exome sequencing was instrumental in making this diagnosis possible. Childhood blindness due to LCA (OMIM 204000) is a severe form of retinal photoreceptor cell degeneration and is defined by the clinical pentad of congenital visual loss, nystagmus (ocular oscillations), amaurotic pupils, retinal degeneration and absent electrical responses from both rods and cones on ERG. Mutations in 15 genes have been associated with autosomal recessive LCA, accounting for ∼70% of the patients. LCA genes are predominantly expressed in the retina, and LCA proteins participate in a variety of retinal pathways. LCA cohort studies have found that the majority of patients have an ocular phenotype only, which correlates with the predominant retinal expression of known LCA genes. However, we recently discovered three ubiquitously expressed ciliary genes that are mutated in LCA and, upon further analyses, are associated with systemic disease. We found that LCA patients with mutations in CEP290 or LCA5 develop olfactory defects. We then identified NPHP5 mutations causing LCA, prompted by protein homology and binding to CEP290 , and found that they may result in kidney failure.1–4 These findings indicate the necessity for a paradigm shift in our understanding …


Investigative Ophthalmology & Visual Science | 2014

A Dominant Mutation in Hexokinase 1 (HK1) Causes Retinitis Pigmentosa

Lori S. Sullivan; Daniel C. Koboldt; Sara J. Bowne; Steven Lang; Susan H. Blanton; Elizabeth L. Cadena; Cheryl E. Avery; Richard Alan Lewis; Kaylie Webb-Jones; Dianna H. Wheaton; David G. Birch; Razck Coussa; Huanan Ren; Irma Lopez; Christina Chakarova; Robert K. Koenekoop; Charles A. Garcia; Robert S. Fulton; Richard Wilson; George M. Weinstock; Stephen P. Daiger

PURPOSE To identify the cause of retinitis pigmentosa (RP) in UTAD003, a large, six-generation Louisiana family with autosomal dominant retinitis pigmentosa (adRP). METHODS A series of strategies, including candidate gene screening, linkage exclusion, genome-wide linkage mapping, and whole-exome next-generation sequencing, was used to identify a mutation in a novel disease gene on chromosome 10q22.1. Probands from an additional 404 retinal degeneration families were subsequently screened for mutations in this gene. RESULTS Exome sequencing in UTAD003 led to identification of a single, novel coding variant (c.2539G>A, p.Glu847Lys) in hexokinase 1 (HK1) present in all affected individuals and absent from normal controls. One affected family member carries two copies of the mutation and has an unusually severe form of disease, consistent with homozygosity for this mutation. Screening of additional adRP probands identified four other families (American, Canadian, and Sicilian) with the same mutation and a similar range of phenotypes. The families share a rare 450-kilobase haplotype containing the mutation, suggesting a founder mutation among otherwise unrelated families. CONCLUSIONS We identified an HK1 mutation in five adRP families. Hexokinase 1 catalyzes phosphorylation of glucose to glucose-6-phosphate. HK1 is expressed in retina, with two abundant isoforms expressed at similar levels. The Glu847Lys mutation is located at a highly conserved position in the protein, outside the catalytic domains. We hypothesize that the effect of this mutation is limited to the retina, as no systemic abnormalities in glycolysis were detected. Prevalence of the HK1 mutation in our cohort of RP families is 1%.


Genetics in Medicine | 2015

Comprehensive analysis of patients with Stargardt macular dystrophy reveals new genotype-phenotype correlations and unexpected diagnostic revisions.

Jacques Zaneveld; Sorath Noorani Siddiqui; Huajin Li; Xia Wang; Hui Wang; Keqing Wang; Hui Li; Huanan Ren; Irma Lopez; Allison Dorfman; Ayesha Khan; Feng Wang; Jason S. Salvo; Violet Gelowani; Yumei Li; Ruifang Sui; Robert K. Koenekoop; Rui Chen

Purpose:Stargardt macular dystrophy (STGD) results in early central vision loss. We sought to explain the genetic cause of STGD in a cohort of 88 patients from three different cultural backgrounds.Methods:Next-generation sequencing using a novel capture panel was used to search for disease-causing mutations. Patients with undetermined causes were clinically reexamined and tested for copy-number variations as well as intronic mutations.Results:We determined the cause of disease in 67% of our patients. Our analysis identified 35 novel ABCA4 alleles. Eleven patients had mutations in genes not previously reported to cause STGD. Finally, 45% of our patients with unsolved causes had single deleterious mutations in ABCA4, a recessive disease gene. No likely pathogenic copy-number variations were identified.Conclusion:This study expands our knowledge of STGD by identifying dozens of novel alleles that cause the disease. The frequency of single mutations in ABCA4 among STGD patients is higher than that among controls, indicating that these mutations contribute to disease. Disease in 11 patients was explained by mutations outside ABCA4, underlining the need to genotype all retinal disease genes to maximize genetic diagnostic rates. Few ABCA4 mutations were observed in our French Canadian patients. This population may contain an unidentified founder mutation. Our results indicate that copy-number variations are unlikely to be a major cause of STGD.Genet Med 17 4, 262–270.


Human Mutation | 2013

Screening of a Large Cohort of Leber Congenital Amaurosis and Retinitis Pigmentosa Patients Identifies Novel LCA5 Mutations and New Genotype–Phenotype Correlations

Donna S. Mackay; Arundhati Dev Borman; Ruifang Sui; L. Ingeborgh van den Born; Eliot L. Berson; Louise Ocaka; Alice E. Davidson; John R. Heckenlively; Kari Branham; Huanan Ren; Irma Lopez; Maleeha Maria; Maleeha Azam; Arjen Henkes; Ellen A.W. Blokland; Sten Andréasson; Elfride De Baere; Jean Bennett; Gerald J. Chader; Wolfgang Berger; Irina Golovleva; Jacquie Greenberg; Anneke I. den Hollander; Caroline C. W. Klaver; B. Jeroen Klevering; Birgit Lorenz; Markus N. Preising; Raj Ramsear; Lisa Roberts; Ronald Roepman

This study was undertaken to investigate the prevalence of sequence variants in LCA5 in patients with Leber congenital amaurosis (LCA), early‐onset retinal dystrophy (EORD), and autosomal recessive retinitis pigmentosa (arRP); to delineate the ocular phenotypes; and to provide an overview of all published LCA5 variants in an online database. Patients underwent standard ophthalmic evaluations after providing informed consent. In selected patients, optical coherence tomography (OCT) and fundus autofluorescence imaging were possible. DNA samples from 797 unrelated patients with LCA and 211 with the various types of retinitis pigmentosa (RP) were screened by Sanger sequence analysis of all LCA5 exons and intron/exon junctions. Some LCA patients were prescreened by APEX technology or selected based on homozygosity mapping. In silico analyses were performed to assess the pathogenicity of the variants. Segregation analysis was performed where possible. Published and novel LCA5 variants were collected, amended for their correct nomenclature, and listed in a Leiden Open Variation Database (LOVD). Sequence analysis identified 18 new probands with 19 different LCA5 variants. Seventeen of the 19 LCA5 variants were novel. Except for two missense variants and one splice site variant, all variants were protein‐truncating mutations. Most patients expressed a severe phenotype, typical of LCA. However, some LCA subjects had better vision and intact inner segment/outer segment (IS/OS) junctions on OCT imaging. In two families with LCA5 variants, the phenotype was more compatible with EORD with affected individuals displaying preserved islands of retinal pigment epithelium. One of the families with a milder phenotype harbored a homozygous splice site mutation; a second family was found to have a combination of a stop mutation and a missense mutation. This is the largest LCA5 study to date. We sequenced 1,008 patients (797 with LCA, 211 with arRP) and identified 18 probands with LCA5 mutations. Mutations in LCA5 are a rare cause of childhood retinal dystrophy accounting for ∼2% of disease in this cohort, and the majority of LCA5 mutations are likely null. The LCA5 protein truncating mutations are predominantly associated with LCA. However, in two families with the milder EORD, the LCA5 gene analysis revealed a homozygous splice site mutation in one and a stop mutation in combination with a missense mutation in a second family, suggesting that this milder phenotype is due to residual function of lebercilin and expanding the currently known phenotypic spectrum to include the milder early onset RP. Some patients have remaining foveal cone structures (intact IS/OS junctions on OCT imaging) and remaining visual acuities, which may bode well for upcoming treatment trials.

Collaboration


Dive into the Huanan Ren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irma Lopez

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar

Ayesha Khan

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rui Chen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hui Wang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xia Wang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sorath Noorani Siddiqui

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar

Feng Wang

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge