Your browser doesn't support javascript.
loading
Gene Set Enrichment Analsyes Identify Pathways Involved in Genetic Risk for Diabetic Retinopathy.
Sobrin, Lucia; Susarla, Gayatri; Stanwyck, Lynn; Rouhana, John M; Li, Ashley; Pollack, Samuela; Igo, Robert P; Jensen, Richard A; Li, Xiaohui; Ng, Maggie C Y; Smith, Albert V; Kuo, Jane Z; Taylor, Kent D; Freedman, Barry I; Bowden, Donald W; Penman, Alan; Chen, Ching J; Craig, Jamie E; Adler, Sharon G; Chew, Emily Y; Cotch, Mary Frances; Yaspan, Brian; Mitchell, Paul; Wang, Jie Jin; Klein, Barbara E K; Wong, Tien Y; Rotter, Jerome I; Burdon, Kathyrn P; Iyengar, Sudha K; Segrè, Ayellet V.
Afiliação
  • Sobrin L; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary. Electronic address: lucia_sobrin@meei.harvard.edu.
  • Susarla G; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary.
  • Stanwyck L; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary.
  • Rouhana JM; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary.
  • Li A; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary.
  • Pollack S; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
  • Igo RP; Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, Ohio.
  • Jensen RA; Cardiovascular Health Research Unit, Department of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington.
  • Li X; Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
  • Ng MCY; Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Vanderbilt Genetics Institute and Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Ten
  • Smith AV; Department of Medicine, University of Iceland, Reykjavík, Iceland.
  • Kuo JZ; Medical Affairs, Ophthalmology, Sun Pharmaceutical Industries, Inc, Princeton, New Jersey.
  • Taylor KD; Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
  • Freedman BI; Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina.
  • Bowden DW; Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
  • Penman A; Department of Preventive Medicine, John D. Bower School of Population Health (A.P.), Department of Ophthalmology.
  • Chen CJ; Department of Preventive Medicine, John D. Bower School of Population Health (A.P.), Department of Ophthalmology.
  • Craig JE; University of Mississippi Medical Center, Jackson, Mississippi, USA, FHMRI Eye & Vision, Flinders University, Bedford Park, SA, Australia.
  • Adler SG; Department of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor-University of California, Torrance, California.
  • Chew EY; Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
  • Cotch MF; Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
  • Yaspan B; Genentech Inc, South San Francisco, California, USA.
  • Mitchell P; Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia.
  • Wang JJ; Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, Australia; Center of Clinician-Scientist Development, Duke-NUS Medical School, Singapore.
  • Klein BEK; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
  • Wong TY; Center of Clinician-Scientist Development, Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.
  • Rotter JI; Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
  • Burdon KP; Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.
  • Iyengar SK; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Population and Quantitative Health Sciences, Case Western University, Cleveland, Ohio.
  • Segrè AV; From the Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
Am J Ophthalmol ; 233: 111-123, 2022 01.
Article em En | MEDLINE | ID: mdl-34166655
ABSTRACT
To identify functionally related genes associated with diabetic retinopathy (DR) risk using gene set enrichment analyses applied to genome-wide association study meta-analyses.

METHODS:

We analyzed DR GWAS meta-analyses performed on 3246 Europeans and 2611 African Americans with type 2 diabetes. Gene sets relevant to 5 key DR pathophysiology processes were investigated tissue injury, vascular events, metabolic events and glial dysregulation, neuronal dysfunction, and inflammation. Keywords relevant to these processes were queried in 4 pathway and ontology databases. Two GSEA methods, Meta-Analysis Gene set Enrichment of variaNT Associations (MAGENTA) and Multi-marker Analysis of GenoMic Annotation (MAGMA), were used. Gene sets were defined to be enriched for gene associations with DR if the P value corrected for multiple testing (Pcorr) was <.05.

RESULTS:

Five gene sets were significantly enriched for numerous modest genetic associations with DR in one method (MAGENTA or MAGMA) and also at least nominally significant (uncorrected P < .05) in the other method. These pathways were regulation of the lipid catabolic process (2-fold enrichment, Pcorr = .014); nitric oxide biosynthesis (1.92-fold enrichment, Pcorr = .022); lipid digestion, mobilization, and transport (1.6-fold enrichment, P = .032); apoptosis (1.53-fold enrichment, P = .041); and retinal ganglion cell degeneration (2-fold enrichment, Pcorr = .049). The interferon gamma (IFNG) gene, previously implicated in DR by protein-protein interactions in our GWAS, was among the top ranked genes in the nitric oxide pathway (best variant P = .0001).

CONCLUSIONS:

These GSEA indicate that variants in genes involved in oxidative stress, lipid transport and catabolism, and cell degeneration are enriched for genes associated with DR risk. NOTE Publication of this article is sponsored by the American Ophthalmological Society.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Diabetes Mellitus Tipo 2 / Retinopatia Diabética Tipo de estudo: Etiology_studies / Prognostic_studies / Risk_factors_studies / Systematic_reviews Limite: Humans Idioma: En Revista: Am J Ophthalmol Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Diabetes Mellitus Tipo 2 / Retinopatia Diabética Tipo de estudo: Etiology_studies / Prognostic_studies / Risk_factors_studies / Systematic_reviews Limite: Humans Idioma: En Revista: Am J Ophthalmol Ano de publicação: 2022 Tipo de documento: Article