1
|
Vaughn AH, Nielsen R. Fast and Accurate Estimation of Selection Coefficients and Allele Histories from Ancient and Modern DNA. Mol Biol Evol 2024; 41:msae156. [PMID: 39078618 PMCID: PMC11321360 DOI: 10.1093/molbev/msae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
We here present CLUES2, a full-likelihood method to infer natural selection from sequence data that is an extension of the method CLUES. We make several substantial improvements to the CLUES method that greatly increases both its applicability and its speed. We add the ability to use ancestral recombination graphs on ancient data as emissions to the underlying hidden Markov model, which enables CLUES2 to use both temporal and linkage information to make estimates of selection coefficients. We also fully implement the ability to estimate distinct selection coefficients in different epochs, which allows for the analysis of changes in selective pressures through time, as well as selection with dominance. In addition, we greatly increase the computational efficiency of CLUES2 over CLUES using several approximations to the forward-backward algorithms and develop a new way to reconstruct historic allele frequencies by integrating over the uncertainty in the estimation of the selection coefficients. We illustrate the accuracy of CLUES2 through extensive simulations and validate the importance sampling framework for integrating over the uncertainty in the inference of gene trees. We also show that CLUES2 is well-calibrated by showing that under the null hypothesis, the distribution of log-likelihood ratios follows a χ2 distribution with the appropriate degrees of freedom. We run CLUES2 on a set of recently published ancient human data from Western Eurasia and test for evidence of changing selection coefficients through time. We find significant evidence of changing selective pressures in several genes correlated with the introduction of agriculture to Europe and the ensuing dietary and demographic shifts of that time. In particular, our analysis supports previous hypotheses of strong selection on lactase persistence during periods of ancient famines and attenuated selection in more modern periods.
Collapse
Affiliation(s)
- Andrew H Vaughn
- Center for Computational Biology, University of California, Berkeley, CA 94720, USA
| | - Rasmus Nielsen
- Departments of Integrative Biology and Statistics, University of California, Berkeley, CA 94720, USA
- Center for GeoGenetics, University of Copenhagen, Copenhagen DK-1350, Denmark
| |
Collapse
|
2
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
3
|
Liang H, Sedillo JC, Schrodi SJ, Ikeda A. Structural variants in linkage disequilibrium with GWAS-significant SNPs. Heliyon 2024; 10:e32053. [PMID: 38882374 PMCID: PMC11177133 DOI: 10.1016/j.heliyon.2024.e32053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
With the recent expansion of structural variant identification in the human genome, understanding the role of these impactful variants in disease architecture is critically important. Currently, a large proportion of genome-wide-significant genome-wide association study (GWAS) single nucleotide polymorphisms (SNPs) are functionally unresolved, raising the possibility that some of these SNPs are associated with disease through linkage disequilibrium with causal structural variants. Hence, understanding the linkage disequilibrium between newly discovered structural variants and statistically significant SNPs may provide a resource for further investigation into disease-associated regions in the genome. Here we present a resource cataloging structural variant-significant SNP pairs in high linkage disequilibrium. The database is composed of (i) SNPs that have exhibited genome-wide significant association with traits, primarily disease phenotypes, (ii) newly released structural variants (SVs), and (iii) linkage disequilibrium values calculated from unphased data. All data files including those detailing SV and GWAS SNP associations and results of GWAS-SNP-SV pairs are available at the SV-SNP LD Database and can be accessed at 'https://github.com/hliang-SchrodiLab/SV_SNPs. Our analysis results represent a useful fine mapping tool for interrogating SVs in linkage disequilibrium with disease-associated SNPs. We anticipate that this resource may play an important role in subsequent studies which investigate incorporating disease causing SVs into disease risk prediction models.
Collapse
Affiliation(s)
- Hao Liang
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Joni C Sedillo
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven J Schrodi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
4
|
Brodzka S, Baszyński J, Rektor K, Hołderna-Bona K, Stanek E, Kurhaluk N, Tkaczenko H, Malukiewicz G, Woźniak A, Kamiński P. Immunogenetic and Environmental Factors in Age-Related Macular Disease. Int J Mol Sci 2024; 25:6567. [PMID: 38928273 PMCID: PMC11203563 DOI: 10.3390/ijms25126567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Age-related macular degeneration (AMD) is a chronic disease, which often develops in older people, but this is not the rule. AMD pathogenesis changes include the anatomical and functional complex. As a result of damage, it occurs, in the retina and macula, among other areas. These changes may lead to partial or total loss of vision. This disease can occur in two clinical forms, i.e., dry (progression is slowly and gradually) and exudative (wet, progression is acute and severe), which usually started as dry form. A coexistence of both forms is possible. AMD etiology is not fully understood. Extensive genetic studies have shown that this disease is multifactorial and that genetic determinants, along with environmental and metabolic-functional factors, are important risk factors. This article reviews the impact of heavy metals, macro- and microelements, and genetic factors on the development of AMD. We present the current state of knowledge about the influence of environmental factors and genetic determinants on the progression of AMD in the confrontation with our own research conducted on the Polish population from Kuyavian-Pomeranian and Lubusz Regions. Our research is concentrated on showing how polluted environments of large agglomerations affects the development of AMD. In addition to confirming heavy metal accumulation, the growth of risk of acute phase factors and polymorphism in the genetic material in AMD development, it will also help in the detection of new markers of this disease. This will lead to a better understanding of the etiology of AMD and will help to establish prevention and early treatment.
Collapse
Affiliation(s)
- Sylwia Brodzka
- Division of Ecology and Environmental Protection, Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-094 Bydgoszcz, Poland; (S.B.); (J.B.); (K.H.-B.); (E.S.)
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, PL 65-516 Zielona Góra, Poland;
| | - Jędrzej Baszyński
- Division of Ecology and Environmental Protection, Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-094 Bydgoszcz, Poland; (S.B.); (J.B.); (K.H.-B.); (E.S.)
| | - Katarzyna Rektor
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, PL 65-516 Zielona Góra, Poland;
| | - Karolina Hołderna-Bona
- Division of Ecology and Environmental Protection, Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-094 Bydgoszcz, Poland; (S.B.); (J.B.); (K.H.-B.); (E.S.)
| | - Emilia Stanek
- Division of Ecology and Environmental Protection, Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-094 Bydgoszcz, Poland; (S.B.); (J.B.); (K.H.-B.); (E.S.)
| | - Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, PL 76-200 Słupsk, Poland; (N.K.); (H.T.)
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, PL 76-200 Słupsk, Poland; (N.K.); (H.T.)
| | - Grażyna Malukiewicz
- Department of Eye Diseases, University Hospital No. 1, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, PL 85-092 Bydgoszcz, Poland;
| | - Piotr Kamiński
- Division of Ecology and Environmental Protection, Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, PL 85-094 Bydgoszcz, Poland; (S.B.); (J.B.); (K.H.-B.); (E.S.)
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, PL 65-516 Zielona Góra, Poland;
| |
Collapse
|
5
|
Zhao N, Hao XN, Huang JM, Song ZM, Tao Y. Crosstalk Between Microglia and Müller Glia in the Age-Related Macular Degeneration: Role and Therapeutic Value of Neuroinflammation. Aging Dis 2024; 15:1132-1154. [PMID: 37728589 PMCID: PMC11081163 DOI: 10.14336/ad.2023.0823-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive neurodegeneration disease that causes photoreceptor demise and vision impairments. In AMD pathogenesis, the primary death of retinal neurons always leads to the activation of resident microglia. The migration of activated microglia to the ongoing retinal lesion and their morphological transformation from branching to ameboid-like are recognized as hallmarks of AMD pathogenesis. Activated microglia send signals to Müller cells and promote them to react correspondingly to damaging stimulus. Müller cells are a type of neuroglia cells that maintain the normal function of retinal neurons, modulating innate inflammatory responses, and stabilize retinal structure. Activated Müller cells can accelerate the progression of AMD by damaging neurons and blood vessels. Therefore, the crosstalk between microglia and Müller cells plays a homeostatic role in maintaining the retinal environment, and this interaction is complicatedly modulated. In particular, the mechanism of mutual regulation between the two glia populations is complex under pathological conditions. This paper reviews recent findings on the crosstalk between microglia and Müller glia during AMD pathology process, with special emphasis on its therapeutic potentials.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiao-Na Hao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jie-Min Huang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Zong-Ming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China.
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
6
|
He W, Han X, Ong JS, Wu Y, Hewitt AW, Mackey DA, Gharahkhani P, MacGregor S. Genome-Wide Meta-analysis Identifies Risk Loci and Improves Disease Prediction of Age-Related Macular Degeneration. Ophthalmology 2024; 131:16-29. [PMID: 37634759 DOI: 10.1016/j.ophtha.2023.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/22/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023] Open
Abstract
PURPOSE To identify age-related macular degeneration (AMD) risk loci and to establish a polygenic prediction model. DESIGN Genome-wide association study (GWAS) and polygenic risk score (PRS) construction. PARTICIPANTS We included 64 885 European patients with AMD and 568 740 control participants (with overlapped samples) in the UK Biobank, Genetic Epidemiology Research on Aging (GERA), International AMD Consortium, FinnGen, and published early AMD GWASs in meta-analyses, as well as 733 European patients with AMD and 20 487 control participants from the Canadian Longitudinal Study on Aging (CLSA) and non-Europeans from the UK Biobank and GERA for polygenic risk score validation. METHODS A multitrait meta-analysis of GWASs comprised 64 885 patients with AMD and 568 740 control participants; the multitrait approach accounted for sample overlap. We constructed a PRS for AMD based on both previously reported as well as unreported AMD loci. We applied the PRS to nonoverlapping data from the CLSA. MAIN OUTCOME MEASURES We identified several single nucleotide polymorphisms associated with AMD and established a PRS for AMD risk prediction. RESULTS We identified 63 AMD risk loci alongside the well-established AMD loci CFH and ARMS2, including 9 loci that were not reported in previous GWASs, some of which previously were linked to other eye diseases such as glaucoma (e.g., HIC1). We applied our PRS to nonoverlapping data from the CLSA. A new PRS was constructed using the PRS method, PRS-CS, and significantly improved the prediction accuracy of AMD risk compared with PRSs from previously published datasets. We further showed that even people who carry all the well-known AMD risk alleles at CFH and ARMS2 vary considerably in their AMD risk (ranging from close to 0 in individuals with low PRS to > 50% in individuals with high PRS). Although our PRS was derived in individuals of European ancestry, the PRS shows potential for predicting risk in people of East Asian, South Asian, and Latino ancestry. CONCLUSIONS Our findings improve the knowledge of the genetic architecture of AMD and help achieve better accuracy in AMD prediction. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Weixiong He
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| | - Xikun Han
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jue-Sheng Ong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yeda Wu
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Alex W Hewitt
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victorian, Australia; School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - David A Mackey
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
7
|
Orozco LD, Owen LA, Hofmann J, Stockwell AD, Tao J, Haller S, Mukundan VT, Clarke C, Lund J, Sridhar A, Mayba O, Barr JL, Zavala RA, Graves EC, Zhang C, Husami N, Finley R, Au E, Lillvis JH, Farkas MH, Shakoor A, Sherva R, Kim IK, Kaminker JS, Townsend MJ, Farrer LA, Yaspan BL, Chen HH, DeAngelis MM. A systems biology approach uncovers novel disease mechanisms in age-related macular degeneration. CELL GENOMICS 2023; 3:100302. [PMID: 37388919 PMCID: PMC10300496 DOI: 10.1016/j.xgen.2023.100302] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/21/2023] [Accepted: 03/22/2023] [Indexed: 07/01/2023]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness, affecting 200 million people worldwide. To identify genes that could be targeted for treatment, we created a molecular atlas at different stages of AMD. Our resource is comprised of RNA sequencing (RNA-seq) and DNA methylation microarrays from bulk macular retinal pigment epithelium (RPE)/choroid of clinically phenotyped normal and AMD donor eyes (n = 85), single-nucleus RNA-seq (164,399 cells), and single-nucleus assay for transposase-accessible chromatin (ATAC)-seq (125,822 cells) from the retina, RPE, and choroid of 6 AMD and 7 control donors. We identified 23 genome-wide significant loci differentially methylated in AMD, over 1,000 differentially expressed genes across different disease stages, and an AMD Müller state distinct from normal or gliosis. Chromatin accessibility peaks in genome-wide association study (GWAS) loci revealed putative causal genes for AMD, including HTRA1 and C6orf223. Our systems biology approach uncovered molecular mechanisms underlying AMD, including regulators of WNT signaling, FRZB and TLE2, as mechanistic players in disease.
Collapse
Affiliation(s)
- Luz D. Orozco
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Leah A. Owen
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
- Department of Population Health Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Jeffrey Hofmann
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Amy D. Stockwell
- Department of Human Genetics, Genentech, South San Francisco, CA 94080, USA
| | - Jianhua Tao
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Susan Haller
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Vineeth T. Mukundan
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Christine Clarke
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Jessica Lund
- Departments of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA 94080, USA
| | - Akshayalakshmi Sridhar
- Department of Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA 94080, USA
| | - Oleg Mayba
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Julie L. Barr
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Rylee A. Zavala
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Elijah C. Graves
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Charles Zhang
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Nadine Husami
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Robert Finley
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - Elizabeth Au
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| | - John H. Lillvis
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Veterans Administration Western New York Healthcare System, Buffalo, NY 14212, USA
| | - Michael H. Farkas
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Veterans Administration Western New York Healthcare System, Buffalo, NY 14212, USA
| | - Akbar Shakoor
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
| | - Richard Sherva
- Department of Medicine, Biomedical Genetics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivana K. Kim
- Retina Service, Massachusetts Eye & Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Joshua S. Kaminker
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Michael J. Townsend
- Department of Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA 94080, USA
| | - Lindsay A. Farrer
- Department of Medicine, Biomedical Genetics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Brian L. Yaspan
- Department of Human Genetics, Genentech, South San Francisco, CA 94080, USA
| | - Hsu-Hsin Chen
- Department of Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA 94080, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
- Department of Population Health Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT 84132, USA
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
- Genetics, Genomics and Bioinformatics Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
8
|
Khachigian LM, Liew G, Teo KYC, Wong TY, Mitchell P. Emerging therapeutic strategies for unmet need in neovascular age-related macular degeneration. J Transl Med 2023; 21:133. [PMID: 36810060 PMCID: PMC9942398 DOI: 10.1186/s12967-023-03937-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is a major cause of visual impairment and blindness. Anti-vascular endothelial growth factor (VEGF) agents, such as ranibizumab, bevacizumab, aflibercept, brolucizumab and faricimab have revolutionized the clinical management of nAMD. However, there remains an unmet clinical need for new and improved therapies for nAMD, since many patients do not respond optimally, may lose response over time or exhibit sub-optimal durability, impacting on real world effectiveness. Evidence is emerging that targeting VEGF-A alone, as most agents have done until recently, may be insufficient and agents that target multiple pathways (e.g., aflibercept, faricimab and others in development) may be more efficacious. This article reviews issues and limitations that have arisen from the use of existing anti-VEGF agents, and argues that the future may lie in multi-targeted therapies including alternative agents and modalities that target both the VEGF ligand/receptor system as well as other pathways.
Collapse
Affiliation(s)
- Levon M. Khachigian
- grid.1005.40000 0004 4902 0432Vascular Biology and Translational Research, Faculty of Medicine and Health, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052 Australia
| | - Gerald Liew
- grid.476921.fCentre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Westmead, Australia
| | - Kelvin Y. C. Teo
- grid.419272.b0000 0000 9960 1711Singapore National Eye Centre and Singapore Eye Research Institute, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
| | - Tien Y. Wong
- grid.419272.b0000 0000 9960 1711Singapore National Eye Centre and Singapore Eye Research Institute, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore ,grid.12527.330000 0001 0662 3178Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Paul Mitchell
- grid.476921.fCentre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Westmead, Australia
| |
Collapse
|
9
|
Dhirachaikulpanich D, Lagger C, Chatsirisupachai K, de Magalhães JP, Paraoan L. Intercellular communication analysis of the human retinal pigment epithelial and choroidal cells predicts pathways associated with aging, cellular senescence and age-related macular degeneration. Front Aging Neurosci 2022; 14:1016293. [PMID: 36408112 PMCID: PMC9669800 DOI: 10.3389/fnagi.2022.1016293] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
The retinal pigment epithelium (RPE) and the choroid are ocular tissues with fundamental roles in supporting neuroretinal function. The pathogenesis of age-related macular degeneration (AMD), a leading cause of irreversible blindness for which aging is the highest risk factor is closely linked with progressive impairment of various functions of these tissues. Cellular senescence, marked by cell cycle arrest and secretion of proinflammatory factors, is known to be associated with aging and has been proposed as a potential driver of AMD. Here, we investigated the role played by intercellular communication in the RPE/choroid within the context of aging, senescence and AMD. We inferred cell–cell interactions in the RPE/choroid by applying CellChat and scDiffCom on a publicly available scRNA-seq dataset from three human donors with and without AMD. We identified age-regulated ligand and receptor genes by using limma on a separate publicly available bulk microarray dataset providing RPE/choroid samples at multiple time points. Cellular senescence was investigated by assigning a score to each cell and each sample of these scRNA-seq and microarray datasets, respectively, based on the expression of key signature genes determined by a previous senescence meta-analysis. We identified VEGF-, BMP-and tenascin-mediated pathways supporting some of the strongest cell–cell interactions between RPE cells, fibroblasts and choroidal endothelial cells and as strong intercellular communication pathways related to both aging and senescence. Their signaling strength was enhanced between subpopulations of cells having high senescence scores. Predominant ligands of these pathways were upregulated with age whereas predominant receptors were downregulated. Globally, we also observed that cells from AMD samples presented slightly bigger senescence scores than normal cells and that the senescence score positively correlated with age in bulk samples (R = 0.26, value of p < 0.01). Hence, our analysis provides novel information on RPE/choroid intercellular communication that gives insights into the connection between aging, senescence and AMD.
Collapse
Affiliation(s)
- Dhanach Dhirachaikulpanich
- Ocular Molecular Biology and Mechanisms of Disease Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Cyril Lagger
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kasit Chatsirisupachai
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: João Pedro de Magalhães,
| | - Luminita Paraoan
- Ocular Molecular Biology and Mechanisms of Disease Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Luminita Paraoan,
| |
Collapse
|
10
|
Cipriani V, Tierney A, Griffiths JR, Zuber V, Sergouniotis PI, Yates JRW, Moore AT, Bishop PN, Clark SJ, Unwin RD. Beyond factor H: The impact of genetic-risk variants for age-related macular degeneration on circulating factor-H-like 1 and factor-H-related protein concentrations. Am J Hum Genet 2021; 108:1385-1400. [PMID: 34260948 PMCID: PMC8387294 DOI: 10.1016/j.ajhg.2021.05.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/27/2021] [Indexed: 01/04/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss; there is strong genetic susceptibility at the complement factor H (CFH) locus. This locus encodes a series of complement regulators: factor H (FH), a splice variant factor-H-like 1 (FHL-1), and five factor-H-related proteins (FHR-1 to FHR-5), all involved in the regulation of complement factor C3b turnover. Little is known about how AMD-associated variants at this locus might influence FHL-1 and FHR protein concentrations. We have used a bespoke targeted mass-spectrometry assay to measure the circulating concentrations of all seven complement regulators and demonstrated elevated concentrations in 352 advanced AMD-affected individuals for all FHR proteins (FHR-1, p = 2.4 × 10-10; FHR-2, p = 6.0 × 10-10; FHR-3, p = 1.5 × 10-5; FHR-4, p = 1.3 × 10-3; FHR-5, p = 1.9 × 10-4) and FHL-1 (p = 4.9 × 10-4) when these individuals were compared to 252 controls, whereas no difference was seen for FH (p = 0.94). Genome-wide association analyses in controls revealed genome-wide-significant signals at the CFH locus for all five FHR proteins, and univariate Mendelian-randomization analyses strongly supported the association of FHR-1, FHR-2, FHR-4, and FHR-5 with AMD susceptibility. These findings provide a strong biochemical explanation for how genetically driven alterations in circulating FHR proteins could be major drivers of AMD and highlight the need for research into FHR protein modulation as a viable therapeutic avenue for AMD.
Collapse
Affiliation(s)
- Valentina Cipriani
- William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, United Kingdom; Moorfields Eye Hospital National Health Service Foundation Trust, London, EC1V 2PD, United Kingdom; UCL Genetics Institute, University College London, London, WC1E 6BT, United Kingdom.
| | - Anna Tierney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, M13 9NY, United Kingdom
| | - John R Griffiths
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, M13 9NY, United Kingdom
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, United Kingdom
| | - Panagiotis I Sergouniotis
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, United Kingdom; Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester University National Health Service Foundation Trust, Manchester, M13 9WL, United Kingdom
| | - John R W Yates
- UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, United Kingdom; Moorfields Eye Hospital National Health Service Foundation Trust, London, EC1V 2PD, United Kingdom; Department of Medical Genetics, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Anthony T Moore
- UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, United Kingdom; Moorfields Eye Hospital National Health Service Foundation Trust, London, EC1V 2PD, United Kingdom; Ophthalmology Department, University of California San Francisco, San Francisco, CA 94143-0730, USA
| | - Paul N Bishop
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, United Kingdom; Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Simon J Clark
- University Eye Clinic, Department for Ophthalmology, Eberhard Karls University of Tübingen, Tübingen, Baden-Württemberg, 72076, Germany; Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Baden-Württemberg, 72076, Germany; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Richard D Unwin
- Stoller Biomarker Discovery Centre and Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, M13 9NQ, United Kingdom.
| |
Collapse
|
11
|
Hall MA, Wallace J, Lucas AM, Bradford Y, Verma SS, Müller-Myhsok B, Passero K, Zhou J, McGuigan J, Jiang B, Pendergrass SA, Zhang Y, Peissig P, Brilliant M, Sleiman P, Hakonarson H, Harley JB, Kiryluk K, Van Steen K, Moore JH, Ritchie MD. Novel EDGE encoding method enhances ability to identify genetic interactions. PLoS Genet 2021; 17:e1009534. [PMID: 34086673 PMCID: PMC8208534 DOI: 10.1371/journal.pgen.1009534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/16/2021] [Accepted: 04/06/2021] [Indexed: 11/26/2022] Open
Abstract
Assumptions are made about the genetic model of single nucleotide polymorphisms (SNPs) when choosing a traditional genetic encoding: additive, dominant, and recessive. Furthermore, SNPs across the genome are unlikely to demonstrate identical genetic models. However, running SNP-SNP interaction analyses with every combination of encodings raises the multiple testing burden. Here, we present a novel and flexible encoding for genetic interactions, the elastic data-driven genetic encoding (EDGE), in which SNPs are assigned a heterozygous value based on the genetic model they demonstrate in a dataset prior to interaction testing. We assessed the power of EDGE to detect genetic interactions using 29 combinations of simulated genetic models and found it outperformed the traditional encoding methods across 10%, 30%, and 50% minor allele frequencies (MAFs). Further, EDGE maintained a low false-positive rate, while additive and dominant encodings demonstrated inflation. We evaluated EDGE and the traditional encodings with genetic data from the Electronic Medical Records and Genomics (eMERGE) Network for five phenotypes: age-related macular degeneration (AMD), age-related cataract, glaucoma, type 2 diabetes (T2D), and resistant hypertension. A multi-encoding genome-wide association study (GWAS) for each phenotype was performed using the traditional encodings, and the top results of the multi-encoding GWAS were considered for SNP-SNP interaction using the traditional encodings and EDGE. EDGE identified a novel SNP-SNP interaction for age-related cataract that no other method identified: rs7787286 (MAF: 0.041; intergenic region of chromosome 7)–rs4695885 (MAF: 0.34; intergenic region of chromosome 4) with a Bonferroni LRT p of 0.018. A SNP-SNP interaction was found in data from the UK Biobank within 25 kb of these SNPs using the recessive encoding: rs60374751 (MAF: 0.030) and rs6843594 (MAF: 0.34) (Bonferroni LRT p: 0.026). We recommend using EDGE to flexibly detect interactions between SNPs exhibiting diverse action. Although traditional genetic encodings are widely implemented in genetics research, including in genome-wide association studies (GWAS) and epistasis, each method makes assumptions that may not reflect the underlying etiology. Here, we introduce a novel encoding method that estimates and assigns an individualized data-driven encoding for each single nucleotide polymorphism (SNP): the elastic data-driven genetic encoding (EDGE). With simulations, we demonstrate that this novel method is more accurate and robust than traditional encoding methods in estimating heterozygous genotype values, reducing the type I error, and detecting SNP-SNP interactions. We further applied the traditional encodings and EDGE to biomedical data from the Electronic Medical Records and Genomics (eMERGE) Network for five phenotypes, and EDGE identified a novel interaction for age-related cataract not detected by traditional methods, which replicated in data from the UK Biobank. EDGE provides an alternative approach to understanding and modeling diverse SNP models and is recommended for studying complex genetics in common human phenotypes.
Collapse
Affiliation(s)
- Molly A. Hall
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Penn State Cancer Institute, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| | - John Wallace
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Anastasia M. Lucas
- Department of Genetics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yuki Bradford
- Department of Genetics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shefali S. Verma
- Department of Genetics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kristin Passero
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jiayan Zhou
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - John McGuigan
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Beibei Jiang
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | | | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Peggy Peissig
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
| | - Murray Brilliant
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
| | - Patrick Sleiman
- Department of Pediatrics, Center for Applied Genomics, Children’s Hospital of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Hakon Hakonarson
- Department of Pediatrics, Center for Applied Genomics, Children’s Hospital of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John B. Harley
- Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- United States Department of Veterans Affairs Medical Center, Cincinnati, Ohio, United States of America
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Kristel Van Steen
- WELBIO, GIGA-R Medical Genomics-BIO3, University of Liège, Liège, Belgium
- Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Jason H. Moore
- Department of Genetics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marylyn D. Ritchie
- Department of Genetics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
12
|
Christopoulos PF, Gjølberg TT, Krüger S, Haraldsen G, Andersen JT, Sundlisæter E. Targeting the Notch Signaling Pathway in Chronic Inflammatory Diseases. Front Immunol 2021; 12:668207. [PMID: 33912195 PMCID: PMC8071949 DOI: 10.3389/fimmu.2021.668207] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The Notch signaling pathway regulates developmental cell-fate decisions and has recently also been linked to inflammatory diseases. Although therapies targeting Notch signaling in inflammation in theory are attractive, their design and implementation have proven difficult, at least partly due to the broad involvement of Notch signaling in regenerative and homeostatic processes. In this review, we summarize the supporting role of Notch signaling in various inflammation-driven diseases, and highlight efforts to intervene with this pathway by targeting Notch ligands and/or receptors with distinct therapeutic strategies, including antibody designs. We discuss this in light of lessons learned from Notch targeting in cancer treatment. Finally, we elaborate on the impact of individual Notch members in inflammation, which may lay the foundation for development of therapeutic strategies in chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Torleif T. Gjølberg
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Eye Research and Department of Ophthalmology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stig Krüger
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eirik Sundlisæter
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Liu X, Song Z, Li Y, Yao Y, Fang M, Bai C, An P, Chen H, Chen Z, Tang B, Shen J, Gao X, Zhang M, Chen P, Zhang T, Jia H, Liu X, Hou Y, Yang H, Wang J, Wang F, Xu X, Min J, Nie C, Zeng Y. Integrated genetic analyses revealed novel human longevity loci and reduced risks of multiple diseases in a cohort study of 15,651 Chinese individuals. Aging Cell 2021; 20:e13323. [PMID: 33657282 PMCID: PMC7963337 DOI: 10.1111/acel.13323] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in studying the genetic contributions to longevity, but limited relevant genes have been identified. In this study, we performed a genetic association study of longevity in a total of 15,651 Chinese individuals. Novel longevity loci, BMPER (rs17169634; p = 7.91 × 10-15 ) and TMEM43/XPC (rs1043943; p = 3.59 × 10-8 ), were identified in a case-control analysis of 11,045 individuals. BRAF (rs1267601; p = 8.33 × 10-15 ) and BMPER (rs17169634; p = 1.45 × 10-10 ) were significantly associated with life expectancy in 12,664 individuals who had survival status records. Additional sex-stratified analyses identified sex-specific longevity genes. Notably, sex-differential associations were identified in two linkage disequilibrium blocks in the TOMM40/APOE region, indicating potential differences during meiosis between males and females. Moreover, polygenic risk scores and Mendelian randomization analyses revealed that longevity was genetically causally correlated with reduced risks of multiple diseases, such as type 2 diabetes, cardiovascular diseases, and arthritis. Finally, we incorporated genetic markers, disease status, and lifestyles to classify longevity or not-longevity groups and predict life span. Our predictive models showed good performance (AUC = 0.86 for longevity classification and explained 19.8% variance of life span) and presented a greater predictive efficiency in females than in males. Taken together, our findings not only shed light on the genetic contributions to longevity but also elucidate correlations between diseases and longevity.
Collapse
Affiliation(s)
- Xiaomin Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- BGI Education Center University of Chinese Academy of Sciences Shenzhen China
| | - Zijun Song
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Yan Li
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yao Yao
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| | - Mingyan Fang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Chen Bai
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
- School of Labor and Human Resources Renmin University Beijing China
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Huashuai Chen
- Business School of Xiangtan University Xiangtan China
| | - Zhihua Chen
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Biyao Tang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Juan Shen
- BGI Genomics BGI‐Shenzhen Shenzhen China
| | - Xiaotong Gao
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | | | - Pengyu Chen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Tao Zhang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huijue Jia
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Xiao Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yong Hou
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huanming Yang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Jian Wang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Fudi Wang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Xun Xu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- Guangdong Provincial Key Laboratory of Genome Read and Write Shenzhen China
| | - Junxia Min
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Chao Nie
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yi Zeng
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| |
Collapse
|
14
|
Comparison of machine learning tools for the prediction of AMD based on genetic, age, and diabetes-related variables in the Chinese population. Regen Ther 2021; 15:180-186. [PMID: 33426217 PMCID: PMC7770346 DOI: 10.1016/j.reth.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Age-related macular degeneration (AMD) is the main cause of visual impairment and the most important cause of blindness in older people. However, there is currently no effective treatment for this disease, so it is necessary to establish a risk model to predict AMD development. Methods This study included a total of 202 subjects, comprising 82 AMD patients and 120 control subjects. Sixty-six single-nucleotide polymorphisms (SNPs) were identified using the MassArray assay. Considering 14 independent clinical variables as well as SNPs, four predictive models were established in the training set and evaluated by the confusion matrix, area under the receiver operating characteristic (ROC) curve (AUROC). The difference distributions of the 14 independent clinical features between the AMD and control groups were tested using the chi-squared test. Age and diabetes were adjusted using logistic regression analysis and the “genomic-control” method was used for multiple testing correction. Results Three SNPs (rs10490924, OR = 1.686, genomic-control corrected p-value (GC) = 0.030; rs2338104, OR = 1.794, GC = 0.025 and rs1864163, OR = 2.125, GC = 0.038) were significant risk factors for AMD development. In the training set, four models obtained AUROC values above 0.72. Conclusions We believe machine learning tools will be useful for the early prediction of AMD and for the development of relevant intervention strategies.
Collapse
|
15
|
Yang C, Wan X, Lin X, Chen M, Zhou X, Liu J. CoMM: a collaborative mixed model to dissecting genetic contributions to complex traits by leveraging regulatory information. Bioinformatics 2020; 35:1644-1652. [PMID: 30295737 DOI: 10.1093/bioinformatics/bty865] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/15/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
MOTIVATION Genome-wide association studies (GWASs) have been successful in identifying many genetic variants associated with complex traits. However, the mechanistic links between these variants and complex traits remain elusive. A scientific hypothesis is that genetic variants influence complex traits at the organismal level via affecting cellular traits, such as regulating gene expression and altering protein abundance. Although earlier works have already presented some scientific insights about this hypothesis and their findings are very promising, statistical methods that effectively harness multilayered data (e.g. genetic variants, cellular traits and organismal traits) on a large scale for functional and mechanistic exploration are highly demanding. RESULTS In this study, we propose a collaborative mixed model (CoMM) to investigate the mechanistic role of associated variants in complex traits. The key idea is built upon the emerging scientific evidence that genetic effects at the cellular level are much stronger than those at the organismal level. Briefly, CoMM combines two models: the first model relating gene expression with genotype and the second model relating phenotype with predicted gene expression using the first model. The two models are fitted jointly in CoMM, such that the uncertainty in predicting gene expression has been fully accounted. To demonstrate the advantages of CoMM over existing methods, we conducted extensive simulation studies, and also applied CoMM to analyze 25 traits in NFBC1966 and Genetic Epidemiology Research on Aging (GERA) studies by integrating transcriptome information from the Genetic European in Health and Disease (GEUVADIS) Project. The results indicate that by leveraging regulatory information, CoMM can effectively improve the power of prioritizing risk variants. Regarding the computational efficiency, CoMM can complete the analysis of NFBC1966 dataset and GERA datasets in 2 and 18 min, respectively. AVAILABILITY AND IMPLEMENTATION The developed R package is available at https://github.com/gordonliu810822/CoMM. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Can Yang
- Department of Mathematics, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiang Wan
- Shenzhen Research Institute of Big Data, Shenzhen, China
| | - Xinyi Lin
- Centre for Quantitative Medicine, Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore
| | - Mengjie Chen
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jin Liu
- Centre for Quantitative Medicine, Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore
| |
Collapse
|
16
|
Fernández‐Vega B, García M, Olivares L, Álvarez L, González‐Fernández A, Artime E, Fernández‐Vega Cueto A, Cobo T, Coca‐Prados M, Vega JA, González‐Iglesias H. The association study of lipid metabolism gene polymorphisms with AMD identifies a protective role for APOE-E2 allele in the wet form in a Northern Spanish population. Acta Ophthalmol 2020; 98:e282-e291. [PMID: 31654486 DOI: 10.1111/aos.14280] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE To elucidate the potential role of eleven single nucleotide polymorphisms (SNPs) in the most relevant lipid metabolism genes in Northern Spanish patients with age-related macular degeneration (AMD). METHODS A case-control study of 228 unrelated native Northern Spanish patients diagnosed with AMD (73 dry and 155 wet) and 95 healthy controls was performed. DNA was isolated from peripheral blood and genotyped for the SNPs APOE rs429358 and rs7412; CTEP rs3764261; LIPC rs10468017 and rs493258; LPL rs12678919; ABCA1 rs1883025; ABCA4 rs76157638, rs3112831 and rs1800555; and SCARB1 rs5888, using TaqMan probes. An additional association study of ε2, ε3 and ε4 major isoforms of APOE gene with AMD has been carried out. RESULTS The allele and genotype frequencies for each of the eleven sequence variants in the lipid metabolism genes did not show significant differences when comparing AMD cases and controls. Statistical analysis revealed that APOE-ε2 carrier genotypes were less frequently observed in patients with wet AMD compared to controls (5.8% versus 13.7%, respectively: p = 3.28 × 10-2 ; OR = 0.42, 95% CI: 0.19-0.95). The frequency of the allele T of rs10468017 (LIPC gene) was lower in dry AMD cases compared to controls (15.8 versus 27.9%, respectively: p = 8.4 × 10-3 OR = 0.57, 95% CI: 0.33-0.98). CONCLUSIONS Our results suggest a protective role for APOE-ε2 allele to wet AMD in the Northern Spanish population.
Collapse
Affiliation(s)
- Beatriz Fernández‐Vega
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
- Departamento de Morfología y Biología Celular Grupo SINPOS Universidad de Oviedo Oviedo Spain
| | - Montserrat García
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Lorena Olivares
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Lydia Álvarez
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Adrián González‐Fernández
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Enol Artime
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Andrés Fernández‐Vega Cueto
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico‐Quirúrgicas Universidad de Oviedo Oviedo Spain
| | - Miguel Coca‐Prados
- Department of Ophthalmology and Visual Science Yale University School of Medicine New Haven CT USA
| | - José A. Vega
- Departamento de Morfología y Biología Celular Grupo SINPOS Universidad de Oviedo Oviedo Spain
- Facultad de Ciencias de la Salud Universidad Autónoma de Chile Santiago de Chile Chile
| | - Héctor González‐Iglesias
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| |
Collapse
|
17
|
Chen LJ. Genetic Association of Age-Related Macular Degeneration and Polypoidal Choroidal Vasculopathy. Asia Pac J Ophthalmol (Phila) 2020; 9:104-109. [PMID: 32195675 DOI: 10.1097/01.apo.0000656976.47696.7d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV) are leading causes of irreversible blindness among the elderly population in developed countries. Although being considered as different subtypes of a same disease, neovascular AMD and PCV have differences in clinical, epidemiological, therapeutic, and genetic profiles. Both AMD and PCV are complex diseases involving multiple genetic and environmental risk factors. Different genetic strategies have been adopted to discover associated genes and variants for neovascular AMD and PCV, including genome-wide association study (GWAS), next-generation sequencing (NGS) based sequence analysis, and candidate gene analyses. So far, a number of susceptible genes have been identified for AMD and/or PCV, such as CFH, ARMS2-HTRA1, C2-CFB-SKIV2L, C3, CETP, and FGD6. Although many of these genes are shared by AMD and PCV, some showed difference between them, such as ARMS2-HTRA1 and FGD6. Also, some of the genes showed ethnic diversities, such as the CFH p.Tyr402His variant. Further larger-scale genomic studies should be warranted to identify more susceptibility genes for AMD and, in particular, PCV among different populations, and differentiate the genetic architectures between neovascular AMD and PCV.
Collapse
Affiliation(s)
- Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital Eye Center, Hong Kong, China
| |
Collapse
|
18
|
A Notch4 missense mutation is associated with susceptibility to tuberculosis in Chinese population. INFECTION GENETICS AND EVOLUTION 2019; 78:104145. [PMID: 31838262 DOI: 10.1016/j.meegid.2019.104145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND The infection process of tuberculosis is related to the interactions between Mycobacterium tuberculosis (MTB) and the host immune system. Polymorphisms in genes involved in the host immune system are related to susceptibility to tuberculosis. The Notch signalling pathway regulates innate and adaptive immunity. Notch4 is a member of the Notch receptor family and may be a negative regulator of Mtb-induced inflammation. However, little is known about the association between Notch4 genetic polymorphisms and susceptibility to tuberculosis; therefore, we explored the association between Notch4 variants and susceptibility to tuberculosis in China. MATERIALS AND METHODS A total of 900 tuberculosis patients and 1534 healthy people serving as controls were enrolled consecutively at West China Hospital between January 2014 and February 2016 Twelve selected SNPs (rs2071277, rs2071285, rs206016, rs438475, rs2256594, rs429853, rs422951, rs415929, rs915895, rs443198, rs3830041 and rs375244) were genotyped by a custom-by-design 2 48-plex SNP scan TM kit. The frequencies of the alleles, genotypes and genetic models of the variants were compared between the two groups, while the SNP-SNP interactions were analysed by Multifactor Dimensionality Reduction (MDR) software. The odds ratio (OR) with a corresponding 95% confidence interval (CI) was calculated. RESULTS The G allele rs2071277 of Notch4 was associated with a decreased risk for tuberculosis (OR 0.844; 95% CI 0.748-0.954, p = .006). The G allele rs422951 of Notch4 was associated with a decreased risk for tuberculosis (OR 0.818; 95% CI 0.703-0.950, p = .008). These findings were consistent with the results from both the dominant model and additive model. The allele, genotype and genetic model frequencies for the other SNPs were similar in the two groups (all P > .05). One haplotype (GTG) consisting of rs2071277, rs2071285 and rs206016 was associated with tuberculosis risk (p = .011). CONCLUSION Ours is the first study implies that the G allele variants of rs2071277 and rs422951 in Notch4 influence susceptibility to tuberculosis in a Chinese population, suggesting that Notch signalling is involved in the pathogenesis of tuberculosis. More studies with functional verification will refine our understanding of the role of Notch signalling and provide novel avenues for therapeutic intervention.
Collapse
|
19
|
Teng MS, Wu S, Hsu LA, Tzeng IS, Chou HH, Su CW, Ko YL. Pleiotropic association of LIPC variants with lipid and urinary 8-hydroxy deoxyguanosine levels in a Taiwanese population. Lipids Health Dis 2019; 18:111. [PMID: 31077211 PMCID: PMC6511151 DOI: 10.1186/s12944-019-1057-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/24/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hepatic lipase (HL, encoded by LIPC) is a glycoprotein primarily synthesized and secreted by hepatocytes. Previous studies had demonstrated that HL is crucial for reverse cholesterol transport and affects the metabolism, composition, and level of several lipoproteins. In current study, we investigated the association of LIPC (Lipase C, Hepatic Type) variants with circulating and urinary biomarker levels by using subgroup and mediation analyses. METHODS A total of 572 participants from Taiwan were genotyped for three LIPC single nucleotide polymorphisms (SNPs) by using TaqMan assay. Fasting levels of glucose, lipid profile, inflammation markers, urine creatinine and 8-hydroxy deoxyguanosine (8-OHdG) were measured. The chi-square test, 2-sample t test and Analysis of variance (ANOVA) were used to examine differences among variables and genotype frequencies. RESULTS SNPs rs2043085 and rs1532085 were significantly associated with urinary 8-OHdG levels, whereas all three SNPs were more significantly associated with Triglycerides (TG) or HDL-cholesterol (HDL-C) levels after additional adjustment for HDL-C or TG levels, respectively. Subgroup analyses revealed that the association of the LIPC SNPs with the levels of serum TG, HDL-C, and urinary 8-OHdG were predominantly observed in the men but not in the women. Differential associations of the LIPC SNPs with various lipid levels were observed in participants with different adiposity statuses. Mediation analyses indicated that TG levels acted as a suppressor masking the association of the LIPC genotypes with HDL-C levels, particularly in the men (Sobel test, all P < 0.01). CONCLUSION Our data revealed that interaction and suppression effects mediated the pleiotropic association of the LIPC variants. The effects of the LIPC SNPs depended on sex, adiposity status, and TG levels. Thus, our findings can provide a method for identifying high-risk populations of cardiovascular diseases for clinical diagnosis.
Collapse
Affiliation(s)
- Ming-Sheng Teng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Semon Wu
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
- Department of Life Science, Chinese Culture University, Taipei, Taiwan
| | - Lung-An Hsu
- The First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Hsin-Hua Chou
- The Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Cheng-Wen Su
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Yu-Lin Ko
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan.
- The Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan.
- School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
20
|
Halladay CW, Hadi T, Anger MD, Greenberg PB, Sullivan JM, Konicki PE, Peachey NS, Igo RP, Iyengar SK, Wu WC, Crawford DC. Genetically-guided algorithm development and sample size optimization for age-related macular degeneration cases and controls in electronic health records from the VA Million Veteran Program. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2019; 2019:153-162. [PMID: 31258967 PMCID: PMC6568141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electronic health records (EHRs) linked to extensive biorepositories and supplemented with lifestyle, behavioral, and environmental exposure data, have enormous potential to contribute to genomic discovery, a necessary step in the pathway towards translational or precision medicine. A major bottleneck in incorporating EHRs into genomic studies is the extraction of research-grade variables for analysis, particularly when gold-standard measurements are not available or accessible. Here we develop algorithms for age-related macular degeneration (AMD), a common cause of blindness among the elderly, and controls free of AMD. These computable phenotypes were developed using billing codes (ICD-9-CM and ICD-10-CM) and Current Procedural Terminology (CPT) codes and evaluated in two study sites of the Veterans Affairs Million Veteran Program: Louis Stokes Cleveland VA Medical Center and the Providence VA Medical Center. After establishing a high overall positive and negative predictive values (93% and 95%, respectively) through manual chart review, the candidate algorithm was deployed in the full VA MVP dataset of >500,000 participants. The algorithm was then optimized in a data cube using a variety of approaches including adjusting inclusion age thresholds by examining previously-reported genetic associations for CFH (rs10801555, a proxy for rs1061170) and ARMS2 (rs10490924). The algorithm with the smallest p-values for the known genetic associations was selected for downstream and on-going AMD genomic discovery efforts. This two-phase approach to developing research-grade case/control variables for AMD genomic studies capitalizes on established genetic associations resulting in high precision and optimized sample sizes, an approach that can be applied to other large-scale biobanks linked to EHRs for precision medicine research.
Collapse
Affiliation(s)
- Christopher W Halladay
- Center for Innovation in Long Term Services and Supports, Providence VA Medical Center, Providence, RI
| | - Tamer Hadi
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH
| | - Matthew D Anger
- Research Service, VA Western NY Healthcare System, Buffalo, NY
- Ophthalmology, SUNY-University at Buffalo, Buffalo, NY
| | - Paul B Greenberg
- Section of Ophthalmology, Providence VA Medical Center, Providence, RI
- Division of Ophthalmology, Alpert Medical School, Brown University, Providence RI
| | - Jack M Sullivan
- Research Service, VA Western NY Healthcare System, Buffalo, NY
- Ophthalmology, SUNY-University at Buffalo, Buffalo, NY
| | - P Eric Konicki
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH
| | - Neal S Peachey
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
| | - Robert P Igo
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| | - Sudha K Iyengar
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH
| | - Wen-Chih Wu
- Section of Cardiology, Medical Service, Providence VA Medical Center, Providence, RI
- Division of Cardiology, Department of Medicine, Alpert Medical School, Brown University, Providence, RI
| | - Dana C Crawford
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH
- Corresponding author
| |
Collapse
|
21
|
Liutkeviciene R, Vilkeviciute A, Kriauciuniene L, Deltuva VP. SIRT1 rs12778366, FGFR2 rs2981582, STAT3 rs744166, LIPC rs10468017, rs493258 and LPL rs12678919 genotypes and haplotype evaluation in patients with age-related macular degeneration. Gene 2019; 686:8-15. [DOI: 10.1016/j.gene.2018.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 10/11/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023]
|
22
|
Porter LF, Saptarshi N, Fang Y, Rathi S, den Hollander AI, de Jong EK, Clark SJ, Bishop PN, Olsen TW, Liloglou T, Chavali VRM, Paraoan L. Whole-genome methylation profiling of the retinal pigment epithelium of individuals with age-related macular degeneration reveals differential methylation of the SKI, GTF2H4, and TNXB genes. Clin Epigenetics 2019; 11:6. [PMID: 30642396 PMCID: PMC6332695 DOI: 10.1186/s13148-019-0608-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
Background Age-related macular degeneration (AMD) is a degenerative disorder of the central retina and the foremost cause of blindness. The retinal pigment epithelium (RPE) is a primary site of disease pathogenesis. The genetic basis of AMD is relatively well understood; however, this knowledge is yet to yield a treatment for the most prevalent non-neovascular disease forms. Therefore, tissue-specific epigenetic mechanisms of gene regulation are of considerable interest in AMD. We aimed to identify differentially methylated genes associated with AMD in the RPE and differentiate local DNA methylation aberrations from global DNA methylation changes, as local DNA methylation changes may be more amenable to therapeutic manipulation. Methods Epigenome-wide association study and targeted gene expression profiling were carried out in RPE cells from eyes of human donors. We performed genome-wide DNA methylation profiling (Illumina 450k BeadChip array) on RPE cells from 44 human donor eyes (25 AMD and 19 normal controls). We validated the findings using bisulfite pyrosequencing in 55 RPE samples (30 AMD and 25 normal controls) including technical (n = 38) and independent replicate samples (n = 17). Long interspersed nucleotide element 1 (LINE-1) analysis was then applied to assess global DNA methylation changes in the RPE. RT-qPCR on independent donor RPE samples was performed to assess gene expression changes. Results Genome-wide DNA methylation profiling identified differential methylation of multiple loci including the SKI proto-oncogene (SKI) (p = 1.18 × 10−9), general transcription factor IIH subunit H4 (GTF2H4) (p = 7.03 × 10−7), and Tenascin X (TNXB) (p = 6.30 × 10−6) genes in AMD. Bisulfite pyrosequencing validated the differentially methylated locus cg18934822 in SKI, and cg22508626 within GTF2H4, and excluded global DNA methylation changes in the RPE in AMD. We further demonstrated the differential expression of SKI, GTF2H4, and TNXB in the RPE of independent AMD donors. Conclusions We report the largest genome-wide methylation analysis of RPE in AMD along with associated gene expression changes to date, for the first-time reaching genome-wide significance, and identified novel targets for functional and future therapeutic intervention studies. The novel differentially methylated genes SKI and GTF2H4 have not been previously associated with AMD, and regulate disease pathways implicated in AMD, including TGF beta signaling (SKI) and transcription-dependent DNA repair mechanisms (GTF2H4). Electronic supplementary material The online version of this article (10.1186/s13148-019-0608-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Louise F Porter
- St Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, UK. .,Department of Eye and Vision Science, William Duncan Building, University of Liverpool, Liverpool, UK.
| | - Neil Saptarshi
- Department of Eye and Vision Science, William Duncan Building, University of Liverpool, Liverpool, UK
| | - Yongxiang Fang
- Centre for Genomic Research, University of Liverpool, Liverpool, UK
| | - Sonika Rathi
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - Anneke I den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eiko K de Jong
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Simon J Clark
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Paul N Bishop
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | | | | - Venkata R M Chavali
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - Luminita Paraoan
- Department of Eye and Vision Science, William Duncan Building, University of Liverpool, Liverpool, UK
| |
Collapse
|
23
|
Zhou X, Li H, Guo S, Wang J, Shi C, Espitia M, Guo X, Wang Q, Liu M, Assassi S, Reveille JD, Mayes MD. Associations of Multiple NOTCH4 Exonic Variants with Systemic Sclerosis. J Rheumatol 2018; 46:184-189. [PMID: 30442821 DOI: 10.3899/jrheum.180094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Findings from previous genome-wide association studies indicated an association of the NOTCH4 gene with systemic sclerosis (SSc). This is a followup study to fine-map exonic variants of NOTCH4 in SSc. METHODS All exons of NOTCH4 were sequenced and analyzed in a total of 1006 patients with SSc and 1004 controls of US white ancestry with the Ion Torrent system. Identified SSc-associated variants were confirmed with Sanger sequencing, and then examined in a Chinese Han cohort consisting of 576 patients with SSc and 574 controls. The NOTCH4 variants were analyzed for association with SSc as a whole and with SSc clinical and autoantibody subtypes with and without the influence of specific HLA-class II alleles that had been previously identified as major genetic factors in SSc. RESULTS A total of 12 SSc-associated and SSc subtype-associated exonic variants of NOTCH4 were identified in the US cohort. Three of them are nonsynonymous single-nucleotide polymorphisms and 1 is a CTG tandem repeat that encodes for a poly-leucine, all of which are located in the NOTCH4 extracellular domain (NECD). Conditional logistic regression analysis on SSc-associated HLA-class II alleles indicated an independent association of the NOTCH4 variants with SSc autoantibody subtypes. Analysis of the Chinese cohort supported a genetic contribution of NOTCH4 to SSc and its subtypes. CONCLUSION Multiple NOTCH4 exonic variants were associated with SSc and/or SSc subtypes. Several of these variants encode nonsynonymous sequence changes occurring in the NECD, which implicates a potentially functional effect of NOTCH4.
Collapse
Affiliation(s)
- Xiaodong Zhou
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China. .,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School.
| | - Hongye Li
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Shicheng Guo
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Jiucun Wang
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Chunhua Shi
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Maribel Espitia
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Xinjian Guo
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Qingwen Wang
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Mengyuan Liu
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Shervin Assassi
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - John D Reveille
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| | - Maureen D Mayes
- From the Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health, Houston, Texas, USA; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai; Department of Rheumatology, Jiangxi People's Hospital, Nanchang; Department of Rheumatology, Peking University-Shenzhen Hospital, Shenzhen; Life Sciences College, Hubei University, Wuhan, China.,X. Zhou, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; H. Li, MD, PhD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; S. Guo, PhD, Human Genetics Center, Division of Biostatistics, The University of Texas School of Public Health; J. Wang, PhD, State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University; C. Shi, MD, Department of Rheumatology, Jiangxi People's Hospital; M. Espitia, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; X. Guo, BS, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; Q. Wang, MD, PhD, Department of Rheumatology, Peking University-Shenzhen Hospital; M. Liu, PhD, Life Sciences College, Hubei University; S. Assassi, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; J.D. Reveille, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School; M.D. Mayes, MD, Division of Rheumatology, Department of Internal Medicine, University of Texas McGovern Medical School
| |
Collapse
|
24
|
Zhang J, Li S, Hu S, Yu J, Xiang Y. Association between genetic variation of complement C3 and the susceptibility to advanced age-related macular degeneration: a meta-analysis. BMC Ophthalmol 2018; 18:274. [PMID: 30352574 PMCID: PMC6199710 DOI: 10.1186/s12886-018-0945-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The purpose of this study is to discuss whether genetic variants (rs2230199, rs1047286, rs2230205, and rs2250656) in the C3 gene account for a significant risk of advanced AMD. METHODS We performed a meta-analysis using electronic databases to search relevant articles. A total of 40 case-control studies from 38 available articles (20,673 cases and 20,025 controls) were included in our study. RESULTS In our meta-analysis, the pooled results showed that the carriage of G allele for rs2230199 and the T allele for rs1047286 had a tendency to the risk of advanced AMD (OR = 1.49, 95% CI = 1.39-1.59, P < 0.001; OR = 1.45, 95% CI = 1.37-1.54, P < 0.001). Moreover, in the subgroup analysis based on ethnicity, rs2230199 and rs1047286 polymorphisms were more likely to be a predictor of response for Caucasian region (OR = 1.48, 95% CI = 1.38-1.59, P < 0.001; OR = 1.45, 95% CI = 1.37-1.54, P < 0.001). Besides, pooled results suggested that the G allele of rs2230199 could confer susceptibility to advanced AMD in Middle East (OR = 1.62, 95% CI = 1.33-1.97, P < 0.001). CONCLUSION In our meta-analysis, C3 genetic polymorphisms unveiled a positive effect on the risk of advanced AMD, especially in Caucasians. Furthermore, numerous well-designed studies with large sample-size are required to validate this conclusion.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Ophthalmology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, NO, 26 Shengli Street, Wuhan, 430014, Hubei Province, China
| | - Shuang Li
- Department of Ophthalmology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, NO, 26 Shengli Street, Wuhan, 430014, Hubei Province, China
| | - Shuqiong Hu
- Department of Ophthalmology, the Jingzhou aier eye hospital, Jingzhou, Hubei Province, China
| | - Jiguo Yu
- Department of Ophthalmology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, NO, 26 Shengli Street, Wuhan, 430014, Hubei Province, China
| | - Yi Xiang
- Department of Ophthalmology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, NO, 26 Shengli Street, Wuhan, 430014, Hubei Province, China.
| |
Collapse
|
25
|
Mitchell P, Liew G, Gopinath B, Wong TY. Age-related macular degeneration. Lancet 2018; 392:1147-1159. [PMID: 30303083 DOI: 10.1016/s0140-6736(18)31550-2] [Citation(s) in RCA: 988] [Impact Index Per Article: 141.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration is a leading cause of visual impairment and severe vision loss. Clinically, it is classified as early-stage (medium-sized drusen and retinal pigmentary changes) to late-stage (neovascular and atrophic). Age-related macular degeneration is a multifactorial disorder, with dysregulation in the complement, lipid, angiogenic, inflammatory, and extracellular matrix pathways implicated in its pathogenesis. More than 50 genetic susceptibility loci have been identified, of which the most important are in the CFH and ARMS2 genes. The major non-genetic risk factors are smoking and low dietary intake of antioxidants (zinc and carotenoids). Progression from early-stage to late-stage disease can be slowed with high-dose zinc and antioxidant vitamin supplements. Intravitreal anti-vascular endothelial growth factor therapy (eg, ranibizumab, aflibercept, or bevacizumab) is highly effective at treating neovascular age-related macular degeneration, and has markedly decreased the prevalence of visual impairment in populations worldwide. Currently, no proven therapies for atrophic disease are available, but several agents are being investigated in clinical trials. Future progress is likely to be from improved efforts in prevention and risk-factor modification, personalised medicine targeting specific pathways, newer anti-vascular endothelial growth factor agents or other agents, and regenerative therapies.
Collapse
Affiliation(s)
- Paul Mitchell
- Centre for Vision Research, Department of Ophthalmology, Westmead Institute for Medical Research, University of Sydney, Australia.
| | - Gerald Liew
- Centre for Vision Research, Department of Ophthalmology, Westmead Institute for Medical Research, University of Sydney, Australia
| | - Bamini Gopinath
- Centre for Vision Research, Department of Ophthalmology, Westmead Institute for Medical Research, University of Sydney, Australia
| | - Tien Y Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-National University of Singapore, Singapore
| |
Collapse
|
26
|
Lu F, Liu S, Hao Q, Liu L, Zhang J, Chen X, Hu W, Huang P. Association Between Complement Factor C2/C3/CFB/CFH Polymorphisms and Age-Related Macular Degeneration: A Meta-Analysis. Genet Test Mol Biomarkers 2018; 22:526-540. [PMID: 30179527 DOI: 10.1089/gtmb.2018.0110] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Several previous studies have assessed the contribution of polymorphisms in genes encoding the complement factors C2/C3/CFB/CFH with the risk of age-related macular degeneration (AMD), however the results have been inconsistent. We conducted a meta-analysis to systematically review the potential association between complement factor polymorphisms and AMD. METHODS Studies that investigated associations between C2 (rs547154 and rs9332739), C3 (rs1047286), CFB (rs4151667 and rs641153), and CFH (rs551397 and rs2274700) polymorphisms and AMD were identified by searching PubMed, EMBASE, Web of Science, and Cochrane Library databases for articles published prior to January 1, 2018. Odds ratios (ORs) and 95% confidence intervals (95% CIs) were calculated to evaluate the association between these polymorphisms and AMD using Stata 12.0 software. Q and I2 statistics were used to evaluate between-study heterogeneity. Publication bias analyses were conducted using Begg's test. We also conducted an ethnic subgroup analysis. RESULTS A total of 53 studies that included data for 53,774 patients and 56,973 healthy controls were evaluated. The pooled ORs for rs551397, rs2274700, rs4151667, rs641153, rs1047286, rs9332739, and rs547154 in the heterozygote model were 0.53 (95% CI: 0.45-0.61), 0.53 (95% CI: 0.40-0.70), 0.54 (95% CI: 0.46-0.63), 0.48 (95% CI: 0.4-0.57), 1.42 (95% CI: 1.22-1.66), 0.5 (95% CI: 0.45-0.56), and 0.52 (95% CI: 0.43-0.62), respectively. CONCLUSION Our findings from this analysis confirmed the protective role of C2/CFB/CFH polymorphisms in the development of AMD, but showed that the single nucleotide polymorphism in C3 was a high-risk factor for AMD. The racial analysis results suggested that the effect of variant alleles was stronger in Caucasians than Asians.
Collapse
Affiliation(s)
- Feiteng Lu
- 1 Department of Biochemistry, College of Medicine, Nanchang University , Nanchang, P.R. China
| | - Shuang Liu
- 1 Department of Biochemistry, College of Medicine, Nanchang University , Nanchang, P.R. China
| | - Qingyun Hao
- 1 Department of Biochemistry, College of Medicine, Nanchang University , Nanchang, P.R. China
| | - Lixia Liu
- 2 Department of Internal Medicine, Youhao District People's Hospital , Yichun, P.R. China
| | - Jing Zhang
- 3 Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University , Nanchang, P.R. China
| | - Xiaolong Chen
- 4 Department of Epidemiology, School of Public Health, Nanchang University , Nanchang, P.R. China
| | - Wang Hu
- 4 Department of Epidemiology, School of Public Health, Nanchang University , Nanchang, P.R. China
| | - Peng Huang
- 4 Department of Epidemiology, School of Public Health, Nanchang University , Nanchang, P.R. China .,5 Jiangxi Province Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University , Nanchang, P.R. China
| |
Collapse
|
27
|
Kennedy AE, Ozbek U, Dorak MT. What has GWAS done for HLA and disease associations? Int J Immunogenet 2018; 44:195-211. [PMID: 28877428 DOI: 10.1111/iji.12332] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/16/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
The major histocompatibility complex (MHC) is located in chromosome 6p21 and contains crucial regulators of immune response, including human leucocyte antigen (HLA) genes, alongside other genes with nonimmunological roles. More recently, a repertoire of noncoding RNA genes, including expressed pseudogenes, has also been identified. The MHC is the most gene dense and most polymorphic part of the human genome. The region exhibits haplotype-specific linkage disequilibrium patterns, contains the strongest cis- and trans-eQTLs/meQTLs in the genome and is known as a hot spot for disease associations. Another layer of complexity is provided to the region by the extreme structural variation and copy number variations. While the HLA-B gene has the highest number of alleles, the HLA-DR/DQ subregion is structurally most variable and shows the highest number of disease associations. Reliance on a single reference sequence has complicated the design, execution and analysis of GWAS for the MHC region and not infrequently, the MHC region has even been excluded from the analysis of GWAS data. Here, we contrast features of the MHC region with the rest of the genome and highlight its complexities, including its functional polymorphisms beyond those determined by single nucleotide polymorphisms or single amino acid residues. One of the several issues with customary GWAS analysis is that it does not address this additional layer of polymorphisms unique to the MHC region. We highlight alternative approaches that may assist with the analysis of GWAS data from the MHC region and unravel associations with all functional polymorphisms beyond single SNPs. We suggest that despite already showing the highest number of disease associations, the true extent of the involvement of the MHC region in disease genetics may not have been uncovered.
Collapse
Affiliation(s)
- A E Kennedy
- Center for Research Strategy, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - U Ozbek
- Department of Population Health Science and Policy, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M T Dorak
- Head of School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, UK
| |
Collapse
|
28
|
Kesselmeier M, Pütter C, Volckmar AL, Baurecht H, Grallert H, Illig T, Ismail K, Ollikainen M, Silén Y, Keski-Rahkonen A, Bulik CM, Collier DA, Zeggini E, Hebebrand J, Scherag A, Hinney A. High-throughput DNA methylation analysis in anorexia nervosa confirms TNXB hypermethylation. World J Biol Psychiatry 2018; 19:187-199. [PMID: 27367046 DOI: 10.1080/15622975.2016.1190033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Patients with anorexia nervosa (AN) are ideally suited to identify differentially methylated genes in response to starvation. METHODS We examined high-throughput DNA methylation derived from whole blood of 47 females with AN, 47 lean females without AN and 100 population-based females to compare AN with both controls. To account for different cell type compositions, we applied two reference-free methods (FastLMM-EWASher, RefFreeEWAS) and searched for consensus CpG sites identified by both methods. We used a validation sample of five monozygotic AN-discordant twin pairs. RESULTS Fifty-one consensus sites were identified in AN vs. lean and 81 in AN vs. population-based comparisons. These sites have not been reported in AN methylation analyses, but for the latter comparison 54/81 sites showed directionally consistent differential methylation effects in the AN-discordant twins. For a single nucleotide polymorphism rs923768 in CSGALNACT1 a nearby site was nominally associated with AN. At the gene level, we confirmed hypermethylated sites at TNXB. We found support for a locus at NR1H3 in the AN vs. lean control comparison, but the methylation direction was opposite to the one previously reported. CONCLUSIONS We confirm genes like TNXB previously described to comprise differentially methylated sites, and highlight further sites that might be specifically involved in AN starvation processes.
Collapse
Affiliation(s)
- Miriam Kesselmeier
- a Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital , Jena , Germany
| | - Carolin Pütter
- b Institute for Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen , Essen , Germany
| | - Anna-Lena Volckmar
- c Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| | - Hansjörg Baurecht
- d Department of Dermatology, Allergology, and Venereology , University Hospital Schleswig-Holstein , Campus Kiel, Kiel , Germany
| | - Harald Grallert
- e Research Unit of Molecular Epidemiology , Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health , Neuherberg , Germany.,f German Center for Diabetes Research , Neuherberg , Germany
| | - Thomas Illig
- e Research Unit of Molecular Epidemiology , Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health , Neuherberg , Germany.,g Hannover Unified Biobank , Hannover Medical School , Hannover , Germany.,h Institute of Human Genetics , Hannover Medical School , Hannover , Germany
| | - Khadeeja Ismail
- i Department of Public Health , University of Helsinki , Helsinki , Finland
| | - Miina Ollikainen
- i Department of Public Health , University of Helsinki , Helsinki , Finland
| | - Yasmina Silén
- i Department of Public Health , University of Helsinki , Helsinki , Finland
| | | | - Cynthia M Bulik
- j Department of Psychiatry , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA.,k Department of Nutrition , The University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - David A Collier
- l Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London , London , UK.,m Eli Lilly and Company, Erl Wood Manor , Windlesham , UK
| | - Eleftheria Zeggini
- n Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus , Hinxton , Cambridge , UK
| | - Johannes Hebebrand
- c Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| | - André Scherag
- a Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital , Jena , Germany
| | - Anke Hinney
- c Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Essen, University of Duisburg-Essen , Essen , Germany
| | | |
Collapse
|
29
|
Ruamviboonsuk P, Tadarati M, Singhanetr P, Wattanapokayakit S, Kunhapan P, Wanitchanon T, Wichukchinda N, Mushiroda T, Akiyama M, Momozawa Y, Kubo M, Mahasirimongkol S. Genome-wide association study of neovascular age-related macular degeneration in the Thai population. J Hum Genet 2017; 62:957-962. [PMID: 28703135 DOI: 10.1038/jhg.2017.72] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 01/14/2023]
Abstract
We performed a genome-wide association study on 377 cases of neovascular age-related macular degeneration (AMD) and 1074 controls to determine the association of previously reported genetic variants associated with neovascular AMD in the Thai population. All patients were of Thai ancestry. We confirmed the association of age-related maculopathy susceptibility 2 (ARMS2) rs10490924 (P=7.38 × 10-17), HTRA1 rs11200638 (P=5.47 × 10-17) and complement factor H gene (CFH) rs800292 (P=2.53 × 10-8) with neovascular AMD, all loci passing the genome-wide significance level (P<5.22 × 10-8). We also found association of the previously reported CFH rs10737680 (P=1.76 × 10-6) locus in the discovery sample. Two loci not previously reported to be associated with neovascular AMD were selected for replication in 222 cases and 623 controls. The loci included LINCO1317 rs6733379 and rs2384550 on chromosome 12. LINCO1317 rs6733379 (P=3.85 × 10-2) remained significantly associated with neovascular AMD after replication. In conclusion, we confirm that ARMS2, HTRA1 and CFH variants are associated with neovascular AMD in the Thai population. Findings from this study also suggest that variants contributing to the susceptibility of neovascular AMD in the Thai population are mostly similar to other Asians with additional local genetic risks that may specifically be identified in Thai population.
Collapse
Affiliation(s)
- Paisan Ruamviboonsuk
- Department of Ophthalmology, Rajavithi Hospital, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Mongkol Tadarati
- Department of Ophthalmology, Rajavithi Hospital, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Panisa Singhanetr
- Department of Ophthalmology, Rajavithi Hospital, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Sukanya Wattanapokayakit
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Punna Kunhapan
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Thanyapat Wanitchanon
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Nuanjun Wichukchinda
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Taisei Mushiroda
- Laboratory of Pharmacogenomics, Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | | | - Michiaki Kubo
- Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Surakameth Mahasirimongkol
- Medical Genetics Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| |
Collapse
|
30
|
Reinhard J, Roll L, Faissner A. Tenascins in Retinal and Optic Nerve Neurodegeneration. Front Integr Neurosci 2017; 11:30. [PMID: 29109681 PMCID: PMC5660115 DOI: 10.3389/fnint.2017.00030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/03/2017] [Indexed: 02/04/2023] Open
Abstract
Tenascins represent key constituents of the extracellular matrix (ECM) with major impact on central nervous system (CNS) development. In this regard, several studies indicate that they play a crucial role in axonal growth and guidance, synaptogenesis and boundary formation. These functions are not only important during development, but also for regeneration under several pathological conditions. Additionally, tenascin-C (Tnc) represents a key modulator of the immune system and inflammatory processes. In the present review article, we focus on the function of Tnc and tenascin-R (Tnr) in the diseased CNS, specifically after retinal and optic nerve damage and degeneration. We summarize the current view on both tenascins in diseases such as glaucoma, retinal ischemia, age-related macular degeneration (AMD) or diabetic retinopathy. In this context, we discuss their expression profile, possible functional relevance, remodeling of the interacting matrisome and tenascin receptors, especially under pathological conditions.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
31
|
Abstract
Background Visual impairment in elderly people is a considerable health problem that significantly affects quality of life of millions worldwide. The magnitude of this issue is becoming more evident with an aging population and an increasing number of older individuals. Objective The objective of this article was to review the clinical and pathological aspects of age-related macular degeneration (AMD), diagnostic tools, and therapeutic modalities presently available or underway for both atrophic and wet forms of the disease. Methods An online review of the PubMed database was performed, searching for the key words. The search was limited to articles published since 1980 to date. Results Several risk factors have been linked to AMD, such as age (>60 years), lifestyle (smoking and diet), and family history. Although the pathogenesis of AMD remains unclear, genetic factors have been implicated in the condition. Treatment for atrophic AMD is mainly close observation, coupled with nutritional supplements such as zinc and antioxidants, whereas treatment of wet AMD is based on targeting choroidal neovascular membranes. Conclusion Identification of modifiable risk factors would improve the possibilities of preventing the progression of AMD. The role of anti-vascular endothelial growth factor (anti-VEGF) agents has transformed the therapeutic approach of the potentially blinding disease “wet AMD” into a more favorable outcome.
Collapse
Affiliation(s)
- Waseem M Al-Zamil
- Department of Ophthalmology, Imam Abdulrahman Bin Faisal University, Al-Khobar, Saudi Arabia
| | - Sanaa A Yassin
- Department of Ophthalmology, Imam Abdulrahman Bin Faisal University, Al-Khobar, Saudi Arabia
| |
Collapse
|
32
|
Tom JA, Reeder J, Forrest WF, Graham RR, Hunkapiller J, Behrens TW, Bhangale TR. Identifying and mitigating batch effects in whole genome sequencing data. BMC Bioinformatics 2017; 18:351. [PMID: 28738841 PMCID: PMC5525370 DOI: 10.1186/s12859-017-1756-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Large sample sets of whole genome sequencing with deep coverage are being generated, however assembling datasets from different sources inevitably introduces batch effects. These batch effects are not well understood and can be due to changes in the sequencing protocol or bioinformatics tools used to process the data. No systematic algorithms or heuristics exist to detect and filter batch effects or remove associations impacted by batch effects in whole genome sequencing data. RESULTS We describe key quality metrics, provide a freely available software package to compute them, and demonstrate that identification of batch effects is aided by principal components analysis of these metrics. To mitigate batch effects, we developed new site-specific filters that identified and removed variants that falsely associated with the phenotype due to batch effect. These include filtering based on: a haplotype based genotype correction, a differential genotype quality test, and removing sites with missing genotype rate greater than 30% after setting genotypes with quality scores less than 20 to missing. This method removed 96.1% of unconfirmed genome-wide significant SNP associations and 97.6% of unconfirmed genome-wide significant indel associations. We performed analyses to demonstrate that: 1) These filters impacted variants known to be disease associated as 2 out of 16 confirmed associations in an AMD candidate SNP analysis were filtered, representing a reduction in power of 12.5%, 2) In the absence of batch effects, these filters removed only a small proportion of variants across the genome (type I error rate of 3%), and 3) in an independent dataset, the method removed 90.2% of unconfirmed genome-wide SNP associations and 89.8% of unconfirmed genome-wide indel associations. CONCLUSIONS Researchers currently do not have effective tools to identify and mitigate batch effects in whole genome sequencing data. We developed and validated methods and filters to address this deficiency.
Collapse
Affiliation(s)
- Jennifer A Tom
- Bioinformatics and Computational Biology Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Jens Reeder
- Bioinformatics and Computational Biology Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - William F Forrest
- Bioinformatics and Computational Biology Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Robert R Graham
- Human Genetics Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Julie Hunkapiller
- Human Genetics Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Timothy W Behrens
- Human Genetics Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Tushar R Bhangale
- Bioinformatics and Computational Biology Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA.,Human Genetics Department, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| |
Collapse
|
33
|
Lynn SA, Keeling E, Munday R, Gabha G, Griffiths H, Lotery AJ, Ratnayaka JA. The complexities underlying age-related macular degeneration: could amyloid beta play an important role? Neural Regen Res 2017; 12:538-548. [PMID: 28553324 PMCID: PMC5436342 DOI: 10.4103/1673-5374.205083] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) causes irreversible loss of central vision for which there is no effective treatment. Incipient pathology is thought to occur in the retina for many years before AMD manifests from midlife onwards to affect a large proportion of the elderly. Although genetic as well as non-genetic/environmental risks are recognized, its complex aetiology makes it difficult to identify susceptibility, or indeed what type of AMD develops or how quickly it progresses in different individuals. Here we summarize the literature describing how the Alzheimer's-linked amyloid beta (Aβ) group of misfolding proteins accumulate in the retina. The discovery of this key driver of Alzheimer's disease in the senescent retina was unexpected and surprising, enabling an altogether different perspective of AMD. We argue that Aβ fundamentally differs from other substances which accumulate in the ageing retina, and discuss our latest findings from a mouse model in which physiological amounts of Aβ were subretinally-injected to recapitulate salient features of early AMD within a short period. Our discoveries as well as those of others suggest the pattern of Aβ accumulation and pathology in donor aged/AMD tissues are closely reproduced in mice, including late-stage AMD phenotypes, which makes them highly attractive to study dynamic aspects of Aβ-mediated retinopathy. Furthermore, we discuss our findings revealing how Aβ behaves at single-cell resolution, and consider the long-term implications for neuroretinal function. We propose Aβ as a key element in switching to a diseased retinal phenotype, which is now being used as a biomarker for late-stage AMD.
Collapse
Affiliation(s)
- Savannah A Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rosie Munday
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Gagandeep Gabha
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Helen Griffiths
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Eye Unit, University Southampton NHS Trust, Southampton, United Kingdom
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
34
|
Cardinale CJ, Li D, Tian L, Connolly JJ, March ME, Hou C, Wang F, Snyder J, Kim CE, Chiavacci RM, Sleiman PM, Burnham JM, Hakonarson H. Association of a rare NOTCH4 coding variant with systemic sclerosis: a family-based whole exome sequencing study. BMC Musculoskelet Disord 2016; 17:462. [PMID: 27829420 PMCID: PMC5103422 DOI: 10.1186/s12891-016-1320-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a rheumatologic disease with a multifactorial etiology. Genome-wide association studies imply a polygenic, complex mode of inheritance with contributions from variation at the human leukocyte antigen locus and non-coding variation at a locus on chromosome 6p21, among other modestly impactful loci. Here we describe an 8-year-old female proband presenting with diffuse cutaneous SSc/scleroderma and a family history of SSc in a grandfather and maternal aunt. METHODS We employed whole exome sequencing (WES) of three members of this family. We examined rare missense, nonsense, splice-altering, and coding indels matching an autosomal dominant inheritance model. We selected one missense variant for Sanger sequencing confirmation based on its predicted impact on gene function and location in a known SSc genetic locus. RESULTS Bioinformatic analysis found eight candidate variants meeting our criteria. We identified a very rare missense variant in the regulatory NODP domain of NOTCH4 located at the 6p21 locus, c.4245G > A:p.Met1415Ile, segregating with the phenotype. This allele has a frequency of 1.83 × 10-5 by the data of the Exome Aggregation Consortium. CONCLUSION This family suggests a novel mechanism of SSc pathogenesis in which a rare and penetrant coding variation can substantially elevate disease risk in contrast to the more modest non-coding variation typically found at this locus. These results suggest that modulation of the NOTCH4 gene might be responsible for the association signal at chromosome 6p21 in SSc.
Collapse
Affiliation(s)
- Christopher J Cardinale
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Dong Li
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Lifeng Tian
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - John J Connolly
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Michael E March
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Cuiping Hou
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Fengxiang Wang
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - James Snyder
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Cecilia E Kim
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Rosetta M Chiavacci
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA
| | - Patrick M Sleiman
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jon M Burnham
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Rheumatology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Pediatric Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd Ste 1216, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Wang Y, Wang M, Zhang X, Nie J, Zhang M, Liu X, Ma L. The Association between LIPC rs493258 Polymorphism and the Susceptibility to Age-Related Macular Degeneration. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13101022. [PMID: 27763569 PMCID: PMC5086761 DOI: 10.3390/ijerph13101022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/25/2016] [Accepted: 10/08/2016] [Indexed: 12/29/2022]
Abstract
The purpose of this study was to evaluate the association of the hepatic lipase (LIPC) rs493258 polymorphism and susceptibility to age-related macular degeneration (AMD). A systematic search in PubMed, EMBASE, and ISI web of science databases was performed to identify eligible published studies without language restrictions up to April 2016. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) in different stages of AMD were estimated under different genetic models using meta-analytic methods. Seven studies comprising 20,559 cases and 17,200 controls met the inclusion criteria and were included in the meta-analysis. The LIPC rs493258 polymorphism showed a significant association with a lower risk of AMD under the allelic model (OR = 0.87, 95% CI = 0.84–0.90). Significant relationships between the variant and AMD were also observed in other genetic models (OR ranging from 0.71 to 0.86, all p < 0.05). Stratified analysis based on ethnicity found that LIPC rs493258 polymorphism had a significant association with the decreased risk of the disease in the Caucasian population, but not in the Asian population. For late AMD, significant associations of the rs493258 polymorphism with a lower risk of this disease were also observed in the allelic genetic model (OR = 0.87, 95% CI = 0.83–0.90). This meta-analysis demonstrates that the T allele in the LIPC rs493258 polymorphism was significantly associated with the risk of any and late AMD. The associations of the locus with early and late AMD risk in various populations need further exploration.
Collapse
Affiliation(s)
- Yafeng Wang
- The First Affiliated Hospital of Xi'an Jiaotong University, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China.
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Mingxu Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Xiaoqing Zhang
- Department of Public Health, Xi'an Medical University, Xi'an 710021, China.
| | - Jing Nie
- School of Humanities, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Ming Zhang
- Department of Internal Medicine, Xi'an Honghui Hospital, Xi'an 710054, China.
| | - Xiaohong Liu
- The First Affiliated Hospital of Xi'an Jiaotong University, College of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Le Ma
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| |
Collapse
|
36
|
Xu H, Chen M. Targeting the complement system for the management of retinal inflammatory and degenerative diseases. Eur J Pharmacol 2016; 787:94-104. [PMID: 26948311 PMCID: PMC5026403 DOI: 10.1016/j.ejphar.2016.03.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/12/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Abstract
The retina, an immune privileged tissue, has specialized immune defense mechanisms against noxious insults that may exist in diseases such as age-related macular degeneration (AMD), diabetic retinopathy (DR), uveoretinitis and glaucoma. The defense system consists of retinal innate immune cells (including microglia, perivascular macrophages, and a small population of dendritic cells) and the complement system. Under normal aging conditions, retinal innate immune cells and the complement system undergo a low-grade activation (parainflammation) which is important for retinal homeostasis. In disease states such as AMD and DR, the parainflammatory response is dysregulated and develops into detrimental chronic inflammation. Complement activation in the retina is an important part of chronic inflammation and may contribute to retinal pathology in these disease states. Here, we review the evidence that supports the role of uncontrolled or dysregulated complement activation in various retinal degenerative and angiogenic conditions. We also discuss current strategies that are used to develop complement-based therapies for retinal diseases such as AMD. The potential benefits of complement inhibition in DR, uveoretinitis and glaucoma are also discussed, as well as the need for further research to better understand the mechanisms of complement-mediated retinal damage in these disease states.
Collapse
Affiliation(s)
- Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK.
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, UK.
| |
Collapse
|
37
|
Sardell RJ, Bailey JNC, Courtenay MD, Whitehead P, Laux RA, Adams LD, Fortun JA, Brantley MA, Kovach JL, Schwartz SG, Agarwal A, Scott WK, Haines JL, Pericak-Vance MA. Whole exome sequencing of extreme age-related macular degeneration phenotypes. Mol Vis 2016; 22:1062-76. [PMID: 27625572 PMCID: PMC5007100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/27/2016] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Demographic, environmental, and genetic risk factors for age-related macular degeneration (AMD) have been identified; however, a substantial portion of the variance in AMD disease risk and heritability remains unexplained. To identify AMD risk variants and generate hypotheses for future studies, we performed whole exome sequencing for 75 individuals whose phenotype was not well predicted by their genotype at known risk loci. We hypothesized that these phenotypically extreme individuals were more likely to carry rare risk or protective variants with large effect sizes. METHODS A genetic risk score was calculated in a case-control set of 864 individuals (467 AMD cases, 397 controls) based on 19 common (≥1% minor allele frequency, MAF) single nucleotide variants previously associated with the risk of advanced AMD in a large meta-analysis of advanced cases and controls. We then selected for sequencing 39 cases with bilateral choroidal neovascularization with the lowest genetic risk scores to detect risk variants and 36 unaffected controls with the highest genetic risk score to detect protective variants. After minimizing the influence of 19 common genetic risk loci on case-control status, we targeted single variants of large effect and the aggregate effect of weaker variants within genes and pathways. Single variant tests were conducted on all variants, while gene-based and pathway analyses were conducted on three subsets of data: 1) rare (≤1% MAF in the European population) stop, splice, or damaging missense variants, 2) all rare variants, and 3) all variants. All analyses controlled for the effects of age and sex. RESULTS No variant, gene, or pathway outside regions known to be associated with risk for advanced AMD reached genome-wide significance. However, we identified several variants with substantial differences in allele frequency between cases and controls with strong additive effects on affection status after controlling for age and sex. Protective effects trending toward significance were detected at two loci identified in single-variant analyses: an intronic variant in FBLN7 (the gene encoding fibulin 7) and at three variants near pyridoxal (pyridoxine, vitamin B6) kinase (PDXK). Aggregate rare-variant analyses suggested evidence for association at ASRGL1, a gene previously linked to photoreceptor cell death, and at BSDC1. In known AMD loci we also identified 29 novel or rare damaging missense or stop/splice variants in our sample of cases and controls. CONCLUSIONS Identified variants and genes may highlight regions important in the pathogenesis of AMD and are key targets for replication.
Collapse
Affiliation(s)
- Rebecca J. Sardell
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jessica N Cooke Bailey
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Monique D. Courtenay
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Patrice Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Reneé A. Laux
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Larry D. Adams
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jorge A. Fortun
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Milam A. Brantley
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN
| | - Jaclyn L. Kovach
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Stephen G. Schwartz
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Anita Agarwal
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN
| | - William K. Scott
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
38
|
Multiallelic copy number variation in the complement component 4A (C4A) gene is associated with late-stage age-related macular degeneration (AMD). J Neuroinflammation 2016; 13:81. [PMID: 27090374 PMCID: PMC4835888 DOI: 10.1186/s12974-016-0548-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/11/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the leading cause of vision loss in Western societies with a strong genetic component. Candidate gene studies as well as genome-wide association studies strongly implicated genetic variations in complement genes to be involved in disease risk. So far, no association of AMD with complement component 4 (C4) was reported probably due to the complex nature of the C4 locus on chromosome 6. METHODS We used multiplex ligation-dependent probe amplification (MLPA) to determine the copy number of the C4 gene as well as of both relevant isoforms, C4A and C4B, and assessed their association with AMD using logistic regression models. RESULTS Here, we report on the analysis of 2645 individuals (1536 probands and 1109 unaffected controls), across three different centers, for multiallelic copy number variation (CNV) at the C4 locus. We find strong statistical significance for association of increased copy number of C4A (OR 0.81 (0.73; 0.89);P = 4.4 × 10(-5)), with the effect most pronounced in individuals over 78 years (OR 0.67 (0.55; 0.81)) and females (OR 0.77 (0.68; 0.87)). Furthermore, this association is independent of known AMD-associated risk variants in the nearby CFB/C2 locus, particularly in females and in individuals over 78 years. CONCLUSIONS Our data strengthen the notion that complement dysregulation plays a crucial role in AMD etiology, an important finding for early intervention strategies and future therapeutics. In addition, for the first time, we provide evidence that multiallelic CNVs are associated with AMD pathology.
Collapse
|
39
|
Wang Q, Zhao HS, Li L. Association between complement factor I gene polymorphisms and the risk of age-related macular degeneration: a Meta-analysis of literature. Int J Ophthalmol 2016; 9:298-305. [PMID: 26949655 DOI: 10.18240/ijo.2016.02.23] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 05/27/2015] [Indexed: 01/11/2023] Open
Abstract
AIM To systematically review the association between complement factors I (CFI) polymorphisms and age-related macular degeneration (AMD) and to explore whether CFI polymorphisms are associated with AMD. METHODS Meta-analysis of articles published from 1995 to January 2015 of articles involved with AMD and polymorphisms of the CFI gene. Eligible data were pooled in a Meta-analysis, analyzing using STATA software (version 12.0), Review Manager (version 5.2) and different models based on the heterogeneity of effect sizes. Egger's test, Begg's rank correlation methods were used to evaluate for publication bias. RESULTS Thirteen articles were eligible, describing two loci polymorphisms of the CFI gene (of which 12 articles focus on rs10033900T>C and 3 articles focus on rs2285714C>T). For rs10033900T>C, the results of our study revealed that having a mutant allele C, TC, CC and TC+CC was associated with a decreased risk of AMD in all population groups studied (C versus T models, OR=0.84, 95%CI: 0.72-0.99, P=0.04; TC versus TT models OR=0.89, 95%CI: 0.88-0.99, P=0.04; CC versus TT models, OR=0.76, 95%CI: 0.60-0.98, P=0.03; TC+CC versus TT models, OR=0.81, 95%CI:0.65-0.99, P=0.04). We found that C allele were related to lower AMD risk in the Caucasian population by subgroup analysis, but there was no association with AMD under the allele and genotypes comparison in Asian studies. For rs2285714 C>T, the TC, TT genotypes contributed to a higher risk of AMD, compared with the CC carriers and TC+CC (OR=1.34, 95%CI: 1.09-1.63, P=0.004; OR=1.50, 95%CI: 1.25-1.80, P<0.0001). CONCLUSION This Meta-analysis suggests that CFI rs10033900T>C and rs2285714C>T polymorphisms may contribute to AMD.
Collapse
Affiliation(s)
- Qin Wang
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Center of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Hai-Sheng Zhao
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Li Li
- Center of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
40
|
Ye Z, Shuai P, Zhai Y, Li F, Jiang L, Lu F, Wen F, Huang L, Zhang D, Liu X, Lin Y, Luo H, Zhang H, Zhu X, Wu Z, Yang Z, Gong B, Shi Y. Associations of 6p21.3 Region with Age-related Macular Degeneration and Polypoidal Choroidal Vasculopathy. Sci Rep 2016; 6:20914. [PMID: 26861912 PMCID: PMC4748259 DOI: 10.1038/srep20914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/14/2016] [Indexed: 12/31/2022] Open
Abstract
Neovascular age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV) are leading causes of blindness in aging populations. This study was conducted to investigate the associations of chromosome 6p21.3 region, including CFB-SKIV2L-TNXB-FKBPL-NOTCH4 genes, with both neovascular AMD and PCV. Six single nucleotide polymorphisms (SNPs) in this region and two known AMD-associated SNPs in CFH (rs800292) and HTRA1 (rs11200638) were genotyped in a Han Chinese cohort composed of 490 neovascular AMD patients, 419 PCV patients and 1316 controls. Among the SNPs, TNXB rs12153855 and FKBPL rs9391734 conferred an increased susceptibility to neovascular AMD (P = 2.8 × 10−4 and 0.001, OR = 1.80 and 1.76, respectively), while SKIV2L exerted a protective effect on neovascular AMD (P = 2.2 × 10−4, OR = 0.49). Rs12153855C and rs9391734A alleles could further increase the susceptibility to AMD in subjects with rs800292, rs11200638 and rs429608 risk alleles. However, only the association of SKIV2L rs429608 remained significant after adjusting for rs800292, rs11200638 and the other 5 SNPs. The protective haplotype AATGAG exhibited significant association with neovascular AMD (permutation P = 0.015, OR = 0.34). None of the SNPs in this region was associated with PCV. Association profiles of 6p21.3 region showed discrepancy between neovascular AMD and PCV, indicating possible molecular and pathological differences between these two retinal disorders.
Collapse
Affiliation(s)
- Zimeng Ye
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Ping Shuai
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Health Management Center, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yaru Zhai
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Department of ophthalmology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Lu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Feng Wen
- Zhongshan Ophthalmic Center, Guangzhou, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Health Management Center, Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiaoqi Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Lin
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Huaichao Luo
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Clinical Medicine Department, Luzhou Medical College, Luzhou, China
| | - Houbin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Zhengzheng Wu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Department of ophthalmology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Bo Gong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, School of Medicine, Sichuan Academy of Medical Sciences &Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, China
| |
Collapse
|
41
|
Ma L, Tang FY, Chu WK, Young AL, Brelen ME, Pang CP, Chen LJ. Association of toll-like receptor 3 polymorphism rs3775291 with age-related macular degeneration: a systematic review and meta-analysis. Sci Rep 2016; 6:19718. [PMID: 26796995 PMCID: PMC4726375 DOI: 10.1038/srep19718] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/17/2015] [Indexed: 11/24/2022] Open
Abstract
Association of a polymorphism rs3775291 in the toll-like receptor 3 (TLR3) gene with age-related macular degeneration (AMD) had been investigated intensively, with variable results across studies. Here we conducted a meta-analysis to verify the effect of rs3775291 on AMD. We searched for genetic association studies published in PubMed, EMBASE and Web of Science from start dates to March 10, 2015. Totally 235 reports were retrieved and 9 studies were included for meta-analysis, involving 7400 cases and 13579 controls. Summary odds ratios (ORs) with 95% confidence intervals (CIs) for alleles and genotypes were estimated. TLR3 rs3775291 was associated with both geographic atrophy (GA) and neovascular AMD (nAMD), with marginally significant pooled-P values. Stratification analysis by ethnicity indicated that rs3775291 was associated with all forms of AMD, GA and nAMD only in Caucasians (OR = 0.87, 0.78 and 0.77, respectively, for the TT genotype) but not in East Asians. However, the associations could not withstand Bonferroni correction. This meta-analysis has thus revealed suggestive evidence for TLR3 rs3775291 as an associated marker for AMD in Caucasians but not in Asians. This SNP may have only a small effect on AMD susceptibility. Further studies in larger samples are warranted to confirm its role.
Collapse
Affiliation(s)
- Li Ma
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Fang Yao Tang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alvin L Young
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Ophthalmology and Visual Sciences, Prince of Wales hospital, Hong Kong, China
| | - Marten E Brelen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Ophthalmology and Visual Sciences, Prince of Wales hospital, Hong Kong, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Ophthalmology and Visual Sciences, Prince of Wales hospital, Hong Kong, China
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Ophthalmology and Visual Sciences, Prince of Wales hospital, Hong Kong, China
| |
Collapse
|
42
|
CYP4F2 (rs2108622) Gene Polymorphism Association with Age-Related Macular Degeneration. Adv Med 2016; 2016:3917916. [PMID: 27652291 PMCID: PMC5019857 DOI: 10.1155/2016/3917916] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/23/2016] [Accepted: 07/26/2016] [Indexed: 12/19/2022] Open
Abstract
Background. Age-related macular degeneration is the leading cause of blindness in elderly individuals where aetiology and pathophysiology of age-related macular degeneration are not absolutely clear. Purpose. To determine the frequency of the genotype of rs2108622 in patients with early and exudative age-related macular degeneration. Methods. The study enrolled 190 patients with early age-related macular degeneration, 181 patients with exudative age-related macular degeneration (eAMD), and a random sample of 210 subjects from the general population (control group). The genotyping of rs2108622 was carried out using the real-time polymerase chain reaction method. Results. The analysis of rs2108622 gene polymorphism did not reveal any differences in the distribution of C/C, C/T, and T/T genotypes between the early AMD group, the eAMD group, and the control group. The CYP4F2 (1347C>T) T/T genotype was more frequent in males with eAMD compared to females (10.2% versus 0.8%; p = 0.0052); also T/T genotype was less frequently present in eAMD females compared to healthy control females (0.8% versus 6.2%; p = 0.027). Conclusion. Rs2108622 gene polymorphism had no predominant effect on the development of early AMD and eAMD. The T/T genotype was more frequent in males with eAMD compared to females and less frequently present in eAMD females compared to healthy females.
Collapse
|
43
|
Wang YF, Han Y, Zhang R, Qin L, Wang MX, Ma L. CETP/LPL/LIPC gene polymorphisms and susceptibility to age-related macular degeneration. Sci Rep 2015; 5:15711. [PMID: 26503844 PMCID: PMC4621603 DOI: 10.1038/srep15711] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/18/2015] [Indexed: 12/17/2022] Open
Abstract
Three high-density lipoprotein (HDL)-related loci have been reported to be associated with age-related macular degeneration (AMD), but the results were inconsistent. In this study, the cholesteryl ester transfer protein (CETP) rs3764261 variant was significantly associated with an increased risk of AMD (odds ratio [OR] = 1.13, 95% confidence interval [CI]: 1.05–1.21, P < 0.001), and the hepatic lipase (LIPC) rs10468017 variant was associated with a significantly decreased risk of AMD (OR = 0.81, CI: 0.76–0.86, P < 0.001). Individuals carrying the lipoprotein lipase (LPL) rs12678919 polymorphism (A → G) had no significant change in the risk of developing AMD (OR = 1.01, CI: 0.92–1.10, P = 0.17). After adjusting for the complement factor H (CFH) gene, both CETP and LPL conferred a significantly increased AMD risk (ORCETP = 1.17, CI: 1.08–1.26, P < 0.001; ORLPL = 1.11, CI: 1.01–1.22, P = 0.02). Subgroup analysis based on ethnicity revealed a significant association between the CETP variant and AMD in both Americans (OR = 1.12, CI: 1.02–1.23, P = 0.01) and Europeans (OR = 1.10, CI: 1.01–1.19, P = 0.011). This meta-analysis revealed that both CETP rs3764261 and LIPC rs10468017 polymorphisms were significantly associated with AMD risk. After adjustment for the CFH gene, CETP/LPL conferred a significantly increased susceptibility to the disease, indicating potential interactions among genes in the complement system and the lipid metabolism pathway.
Collapse
Affiliation(s)
- Ya-Feng Wang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Yue Han
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Rui Zhang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Li Qin
- The First Affiliated Hospital, Xi'an Jiao tong University College of Medicine, Xi'an, China
| | - Ming-Xu Wang
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| | - Le Ma
- School of Public Health, Xi'an Jiao tong University Health Science Center, Xi'an, China
| |
Collapse
|
44
|
ALTOBELLI EMMA, MARZIONI DANIELA, LATTANZI AMEDEO, ANGELETTI PAOLOMATTEO. HtrA1: Its future potential as a novel biomarker for cancer. Oncol Rep 2015; 34:555-66. [PMID: 26035313 PMCID: PMC4487665 DOI: 10.3892/or.2015.4016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/16/2015] [Indexed: 12/23/2022] Open
Abstract
HtrA1 appears to be involved in several physiological processes as well as in the pathogenesis of conditions such as Alzheimer's disease and osteoarthritis. It has also been hypothesized to play a role as a tumor suppressor. This manuscript reviews the current cancer-related HtrA1 research from the methodological and clinical standpoints including studies regarding its potential role as a tumor marker and/or prognostic factor. PRISMA method was used for study selection. The articles thus collected were examined and selected by two independent reviewers; any disagreement was resolved by a methodologist. A laboratory researcher reviewed the methods and laboratory techniques. Fifteen studies met the inclusion criteria and concerned the following cancer sites: the nervous system, bladder, breast, esophagus, stomach, liver, endometrium, thyroid, ovaries, pleura, lung and skin. Most articles described in vivo studies using a morphological approach and immunohistochemistry, whereas protein expression was quantified as staining intensity scored by two raters. Often the results were not comparable due to the different rating scales and study design. Current research on HtrA1 does not conclusively support its role as a tumor suppressor.
Collapse
Affiliation(s)
- EMMA ALTOBELLI
- Department of Life, Health and Environmental Sciences, Epidemiology and Biostatistics Unit, AUSL Teramo, University of L’Aquila, L’Aquila, Italy
| | - DANIELA MARZIONI
- Department of Experimental and Clinical Medicine, University of Ancona, Ancona, Italy
| | - AMEDEO LATTANZI
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - PAOLO MATTEO ANGELETTI
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
45
|
Osthoff M, Dean MM, Baird PN, Richardson AJ, Daniell M, Guymer RH, Eisen DP. Association Study of Mannose-Binding Lectin Levels and Genetic Variants in Lectin Pathway Proteins with Susceptibility to Age-Related Macular Degeneration: A Case-Control Study. PLoS One 2015; 10:e0134107. [PMID: 26207622 PMCID: PMC4514807 DOI: 10.1371/journal.pone.0134107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/06/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In age-related macular degeneration (AMD) the complement system is thought to be activated by chronic oxidative damage with genetic variants identified in the alternative pathway as susceptibility factors. However, the involvement of the lectin pathway of complement, a key mediator of oxidative damage, is controversial. This study investigated whether mannose-binding lectin (MBL) levels and genetic variants in lectin pathway proteins, are associated with the predisposition to and severity of AMD. METHODS MBL levels and single nucleotide polymorphisms (SNPs) in the MBL2 and the ficolin-2 (FCN2) gene were determined in 109 patients with AMD and 109 age- and sex-matched controls. RESULTS MBL expression levels were equally distributed in both cases (early and late AMD) and controls (p>0.05). However, there was a trend towards higher median MBL levels in cases with late AMD compared to cases with early AMD (1.0 vs. 0.4 μg/ml, p = 0.09) and MBL deficiency (<0.5 μg/ml) was encountered less frequently in the late AMD group (35% vs 56%, p = 0.03). FCN2 and MBL2 allele frequencies were similarly distributed in early and late AMD cases compared with controls (p>0.05 for all analyses) as were MBL2 genotypes. Similarly, there was no significant difference in allele frequencies in any SNPs in either the MBL2 or FCN2 gene in cases with early vs. late AMD. CONCLUSIONS SNPs of lectin pathway proteins investigated in this study were not associated with AMD or AMD severity. However, MBL levels deserve further study in a larger cohort of early vs. late AMD patients to elucidate any real effect on AMD severity.
Collapse
Affiliation(s)
- Michael Osthoff
- Victorian Infectious Diseases Service at the Doherty Institute, Melbourne Health, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Melinda M. Dean
- Research and Development, Australian Red Cross Blood Service, Kelvin Grove, Queensland, Australia
| | - Paul N. Baird
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Andrea J. Richardson
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Mark Daniell
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Robyn H. Guymer
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Damon P. Eisen
- Victorian Infectious Diseases Service at the Doherty Institute, Melbourne Health, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Butler JM, Sharif U, Ali M, McKibbin M, Thompson JP, Gale R, Yang YC, Inglehearn C, Paraoan L. A missense variant in CST3 exerts a recessive effect on susceptibility to age-related macular degeneration resembling its association with Alzheimer's disease. Hum Genet 2015; 134:705-15. [PMID: 25893795 PMCID: PMC4460273 DOI: 10.1007/s00439-015-1552-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/05/2015] [Indexed: 12/27/2022]
Abstract
Age-related macular degeneration (AMD) and Alzheimer's disease (AD) are degenerative, multifactorial diseases involving age-related accumulation of extracellular deposits linked to dysregulation of protein homeostasis. Here, we strengthen the evidence that an nsSNP (p.Ala25Thr) in the cysteine proteinase inhibitor cystatin C gene CST3, previously confirmed by meta-analysis to be associated with AD, is associated with exudative AMD. To our knowledge, this is the first report highlighting a genetic variant that increases the risk of developing both AD and AMD. Furthermore, we demonstrate that the risk associated with the mutant allele follows a recessive model for both diseases. We perform an AMD-CST3 case-control study genotyping 350 exudative AMD Caucasian individuals. Bringing together our data with the previously reported AMD-CST3 association study, the evidence of a recessive effect on AMD risk is strengthened (OR = 1.89, P = 0.005). This effect closely resembles the AD-CST3 recessive effect (OR = 1.73, P = 0.005) previously established by meta-analysis. This resemblance is substantiated by the high correlation between CST3 genotype and effect size across the two diseases (R(2) = 0.978). A recessive effect is in line with the known function of cystatin C, a potent enzyme inhibitor. Its potency means that, in heterozygous individuals, a single functional allele is sufficient to maintain its inhibitory function; only homozygous individuals will lack this form of proteolytic regulation. Our findings support the hypothesis that recessively acting variants account for some of the missing heritability of multifactorial diseases. Replacement therapy represents a translational opportunity for individuals homozygous for the mutant allele.
Collapse
Affiliation(s)
- Joe M. Butler
- />Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L69 3GA UK
| | - Umar Sharif
- />Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L69 3GA UK
| | - Manir Ali
- />Ophthalmology and Neuroscience, University of Leeds, Leeds, LS9 7TF UK
| | - Martin McKibbin
- />Ophthalmology Department, St James’s University Hospital, Leeds, LS9 7TF UK
| | - Joseph P. Thompson
- />Ophthalmology and Neuroscience, University of Leeds, Leeds, LS9 7TF UK
| | - Richard Gale
- />Ophthalmology Department, The York Hospital, York, YO31 8HE UK
| | - Yit C. Yang
- />Ophthalmology, The Royal Wolverhampton NHS Trust, Wolverhampton, WV10 0QP UK
| | - Chris Inglehearn
- />Ophthalmology and Neuroscience, University of Leeds, Leeds, LS9 7TF UK
| | - Luminita Paraoan
- />Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L69 3GA UK
| |
Collapse
|
47
|
Jiang DK, Ma XP, Yu H, Cao G, Ding DL, Chen H, Huang HX, Gao YZ, Wu XP, Long XD, Zhang H, Zhang Y, Gao Y, Chen TY, Ren WH, Zhang P, Shi Z, Jiang W, Wan B, Saiyin H, Yin J, Zhou YF, Zhai Y, Lu PX, Zhang H, Gu X, Tan A, Wang JB, Zuo XB, Sun LD, Liu JO, Yi Q, Mo Z, Zhou G, Liu Y, Sun J, Shugart YY, Zheng SL, Zhang XJ, Xu J, Yu L. Genetic variants in five novel loci including CFB and CD40 predispose to chronic hepatitis B. Hepatology 2015; 62:118-128. [PMID: 25802187 DOI: 10.1002/hep.27794] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
UNLABELLED Hepatitis B virus affects more than 2 billion people worldwide, 350 million of which have developed chronic hepatitis B (CHB). The genetic factors that confer CHB risk are still largely unknown. We sought to identify genetic variants for CHB susceptibility in the Chinese population. We undertook a genome-wide association study (GWAS) in 2,514 CHB cases and 1,130 normal controls from eastern China. We replicated 33 of the most promising signals and eight previously reported CHB risk loci through a two-stage validation totaling 6,600 CHB cases and 8,127 controls in four independent populations, of which two populations were recruited from eastern China, one from northern China and one from southern China. The joint analyses of 9,114 CHB cases and 9,257 controls revealed significant association of CHB risk with five novel loci. Four loci are located in the human leukocyte antigen (HLA) region at 6p21.3, including two nonsynonymous variants (rs12614 [R32W] in complement factor B [CFB], Pmeta =1.28 × 10(-34) ; and rs422951 [T320A] in NOTCH4, Pmeta = 5.33 × 10(-16) ); one synonymous variant (rs378352 in HLA-DOA corresponding to HLA-DOA*010101, Pmeta = 1.04 × 10(-23) ); and one noncoding variant (rs2853953 near HLA-C, Pmeta = 5.06 × 10(-20) ). Another locus is located at 20q13.1 (rs1883832 in the Kozak sequence of CD40, Pmeta = 2.95 × 10(-15) ). Additionally, we validated seven of eight previously reported CHB susceptibility loci (rs3130542 at HLA-C, rs1419881 at TCF19, rs652888 at EHMT2, rs2856718 at HLA-DQB1, rs7453920 at HLA-DQB2, rs3077 at HLA-DPA1, and rs9277535 at HLA-DPA2, which are all located in the HLA region, 9.84 × 10(-71) ≤ Pmeta ≤ 9.92 × 10(-7) ). CONCLUSION Our GWAS identified five novel susceptibility loci for CHB. These findings improve the understanding of CHB etiology and may provide new targets for prevention and treatment of this disease.
Collapse
Affiliation(s)
- De-Ke Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Xiao-Pin Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hongjie Yu
- James D. Watson Institute of Genome Sciences, College of life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangwen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Dong-Lin Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Haitao Chen
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Hui-Xing Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yu-Zhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Xiao-Pan Wu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xi-Dai Long
- Department of Pathology, Youjiang Medical College for Nationalities, Guangxi, China
| | - Hongxing Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Youjie Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Yong Gao
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Tao-Yang Chen
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Wei-Hua Ren
- Luoyang Central Hospital, Affiliated to Zhengzhou University, Luoyang, China
| | - Pengyin Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Zhuqing Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Wei Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Bo Wan
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jianhua Yin
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Yuan-Feng Zhou
- Department of Pathology, Youjiang Medical College for Nationalities, Guangxi, China
| | - Yun Zhai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Pei-Xin Lu
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Hongwei Zhang
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Xiaoli Gu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Aihua Tan
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Jin-Bing Wang
- Qidong Liver Cancer Institute, Qidong People's Hospital, Qidong, China
| | - Xian-Bo Zuo
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China
- State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Liang-Dan Sun
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China
- State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Qing Yi
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
- Division of Cancer Medicine, and the Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zengnan Mo
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ying Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jielin Sun
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - S Lilly Zheng
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL
| | - Xue-Jun Zhang
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China
- State Key Laboratory Incubation Base of Dermatology, Ministry of National Science and Technology, Hefei, China
| | - Jianfeng Xu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute of Biomedical Science, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Cipriani V, Quartilho A, Bunce C, Freemantle N, Doré CJ. Ophthalmic statistics note 7: multiple hypothesis testing—to adjust or not to adjust. Br J Ophthalmol 2015; 99:1155-7. [PMID: 26112870 DOI: 10.1136/bjophthalmol-2015-306784] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Valentina Cipriani
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK UCL Genetics Institute, London, UK
| | - Ana Quartilho
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Catey Bunce
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK London School of Hygiene & Tropical Medicine, London, UK
| | - Nick Freemantle
- Department of Primary Care and Population Health, University College London, London, UK
| | - Caroline J Doré
- UCL Comprehensive Clinical Trials Unit, University College London, London, UK
| | | |
Collapse
|
49
|
Radeke MJ, Radeke CM, Shih YH, Hu J, Bok D, Johnson LV, Coffey PJ. Restoration of mesenchymal retinal pigmented epithelial cells by TGFβ pathway inhibitors: implications for age-related macular degeneration. Genome Med 2015; 7:58. [PMID: 26150894 PMCID: PMC4491894 DOI: 10.1186/s13073-015-0183-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/11/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a leading cause of blindness. Most vision loss occurs following the transition from a disease of deposit formation and inflammation to a disease of neovascular fibrosis and/or cell death. Here, we investigate how repeated wound stimulus leads to seminal changes in gene expression and the onset of a perpetual state of stimulus-independent wound response in retinal pigmented epithelial (RPE) cells, a cell-type central to the etiology of AMD. METHODS Transcriptome wide expression profiles of human fetal RPE cell cultures as a function of passage and time post-plating were determined using Agilent 44 K whole genome microarrays and RNA-Seq. Using a systems level analysis, differentially expressed genes and pathways of interest were identified and their role in the establishment of a persistent mesenchymal state was assessed using pharmacological-based experiments. RESULTS Using a human fetal RPE cell culture model that considers monolayer disruption and subconfluent culture as a proxy for wound stimulus, we show that prolonged wound stimulus leads to terminal acquisition of a mesenchymal phenotype post-confluence and altered expression of more than 40 % of the transcriptome. In contrast, at subconfluence fewer than 5 % of expressed transcripts have two-fold or greater expression differences after repeated passage. Protein-protein and pathway interaction analysis of the genes with passage-dependent expression levels in subconfluent cultures reveals a 158-node interactome comprised of two interconnected modules with functions pertaining to wound response and cell division. Among the wound response genes are the TGFβ pathway activators: TGFB1, TGFB2, INHBA, INHBB, GDF6, CTGF, and THBS1. Significantly, inhibition of TGFBR1/ACVR1B mediated signaling using receptor kinase inhibitors both forestalls and largely reverses the passage-dependent loss of epithelial potential; thus extending the effective lifespan by at least four passages. Moreover, a disproportionate number of RPE wound response genes have altered expression in neovascular and geographic AMD, including key members of the TGFβ pathway. CONCLUSIONS In RPE cells the switch to a persistent mesenchymal state following prolonged wound stimulus is driven by lasting activation of the TGFβ pathway. Targeted inhibition of TGFβ signaling may be an effective approach towards retarding AMD progression and producing RPE cells in quantity for research and cell-based therapies.
Collapse
Affiliation(s)
- Monte J. Radeke
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Carolyn M. Radeke
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Ying-Hsuan Shih
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Jane Hu
- />Departments of Ophthalmology and Neurobiology, Jules Stein Eye & Brain Research Institutes, David Geffen School of Medicine, University of California, Los Angeles, CA USA
| | - Dean Bok
- />Departments of Ophthalmology and Neurobiology, Jules Stein Eye & Brain Research Institutes, David Geffen School of Medicine, University of California, Los Angeles, CA USA
| | - Lincoln V. Johnson
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| | - Pete J. Coffey
- />Neuroscience Research Institute, University of California, Santa Barbara, CA USA
| |
Collapse
|
50
|
Ratnayaka JA, Serpell LC, Lotery AJ. Dementia of the eye: the role of amyloid beta in retinal degeneration. Eye (Lond) 2015; 29:1013-26. [PMID: 26088679 PMCID: PMC4541342 DOI: 10.1038/eye.2015.100] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/23/2015] [Indexed: 11/09/2022] Open
Abstract
Age-related macular degeneration (AMD) is one of the most common causes of irreversible blindness affecting nearly 50 million individuals globally. The disease is characterised by progressive loss of central vision, which has significant implications for quality of life concerns in an increasingly ageing population. AMD pathology manifests in the macula, a specialised region of the retina, which is responsible for central vision and perception of fine details. The underlying pathology of this complex degenerative disease is incompletely understood but includes both genetic as well as epigenetic risk factors. The recent discovery that amyloid beta (Aβ), a highly toxic and aggregate-prone family of peptides, is elevated in the ageing retina and is associated with AMD has opened up new perspectives on the aetiology of this debilitating blinding disease. Multiple studies now link Aβ with key stages of AMD progression, which is both exciting and potentially insightful, as this identifies a well-established toxic agent that aggressively targets cells in degenerative brains. Here, we review the most recent findings supporting the hypothesis that Aβ may be a key factor in AMD pathology. We describe how multiple Aβ reservoirs, now reported in the ageing eye, may target the cellular physiology of the retina as well as associated layers, and propose a mechanistic pathway of Aβ-mediated degenerative change leading to AMD.
Collapse
Affiliation(s)
- J A Ratnayaka
- Clinical and Experimental Science, Faculty of Medicine, University of Southampton, Southampton, UK
| | - L C Serpell
- School of Life Sciences (Biochemistry, Dementia Research Group), University of Sussex, Brighton, UK
| | - A J Lotery
- Clinical and Experimental Science, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|