Your browser doesn't support javascript.
loading
Human genetic variation in GLS2 is associated with development of complicated Staphylococcus aureus bacteremia.
Scott, William K; Medie, Felix Mba; Ruffin, Felicia; Sharma-Kuinkel, Batu K; Cyr, Derek D; Guo, Shengru; Dykxhoorn, Derek M; Skov, Robert L; Bruun, Niels E; Dahl, Anders; Lerche, Christian J; Petersen, Andreas; Larsen, Anders Rhod; Lauridsen, Trine Kiilerich; Johansen, Helle Krogh; Ullum, Henrik; Sørensen, Erik; Hassager, Christian; Bundgaard, Henning; Schønheyder, Henrik C; Torp-Pedersen, Christian; Østergaard, Louise Bruun; Arpi, Magnus; Rosenvinge, Flemming; Erikstrup, Lise T; Chehri, Mahtab; Søgaard, Peter; Andersen, Paal S; Fowler, Vance G.
Afiliación
  • Scott WK; John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America.
  • Medie FM; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States of America.
  • Ruffin F; Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America.
  • Sharma-Kuinkel BK; Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America.
  • Cyr DD; Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America.
  • Guo S; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, United States of America.
  • Dykxhoorn DM; John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America.
  • Skov RL; John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America.
  • Bruun NE; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States of America.
  • Dahl A; Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark.
  • Lerche CJ; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark.
  • Petersen A; Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
  • Larsen AR; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark.
  • Lauridsen TK; Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
  • Johansen HK; Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark.
  • Ullum H; Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark.
  • Sørensen E; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark.
  • Hassager C; Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
  • Bundgaard H; Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
  • Schønheyder HC; Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
  • Torp-Pedersen C; Department of Cardiology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
  • Østergaard LB; Department of Cardiology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
  • Arpi M; Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark.
  • Rosenvinge F; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
  • Erikstrup LT; Clinical Institute, Aalborg University, Aalborg, Denmark.
  • Chehri M; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark.
  • Søgaard P; Clinical Institute, Aalborg University, Aalborg, Denmark.
  • Andersen PS; Department of Clinical Microbiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark.
  • Fowler VG; Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark.
PLoS Genet ; 14(10): e1007667, 2018 10.
Article en En | MEDLINE | ID: mdl-30289878
ABSTRACT
The role of host genetic variation in the development of complicated Staphylococcus aureus bacteremia (SAB) is poorly understood. We used whole exome sequencing (WES) to examine the cumulative effect of coding variants in each gene on risk of complicated SAB in a discovery sample of 168 SAB cases (84 complicated and 84 uncomplicated, frequency matched by age, sex, and bacterial clonal complex [CC]), and then evaluated the most significantly associated genes in a replication sample of 240 SAB cases (122 complicated and 118 uncomplicated, frequency matched for age, sex, and CC) using targeted sequence capture. In the discovery sample, gene-based analysis using the SKAT-O program identified 334 genes associated with complicated SAB at p<3.5 x 10-3. These, along with eight biologically relevant candidate genes were examined in the replication sample. Gene-based analysis of the 342 genes in the replication sample using SKAT-O identified one gene, GLS2, significantly associated with complicated SAB (p = 1.2 x 10-4) after Bonferroni correction. In Firth-bias corrected logistic regression analysis of individual variants, the strongest association across all 10,931 variants in the replication sample was with rs2657878 in GLS2 (p = 5 x 10-4). This variant is strongly correlated with a missense variant (rs2657879, p = 4.4 x 10-3) in which the minor allele (associated here with complicated SAB) has been previously associated with lower plasma concentration of glutamine. In a microarray-based gene-expression analysis, individuals with SAB exhibited significantly lower expression levels of GLS2 than healthy controls. Similarly, Gls2 expression is lower in response to S. aureus exposure in mouse RAW 264.7 macrophage cells. Compared to wild-type cells, RAW 264.7 cells with Gls2 silenced by CRISPR-Cas9 genome editing have decreased IL1-ß transcription and increased nitric oxide production after S. aureus exposure. GLS2 is an interesting candidate gene for complicated SAB due to its role in regulating glutamine metabolism, a key factor in leukocyte activation.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Infecciones Estafilocócicas / Glutaminasa Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Límite: Adolescent / Adult / Aged / Aged80 / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: PLoS Genet Asunto de la revista: GENETICA Año: 2018 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Infecciones Estafilocócicas / Glutaminasa Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Límite: Adolescent / Adult / Aged / Aged80 / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: PLoS Genet Asunto de la revista: GENETICA Año: 2018 Tipo del documento: Article País de afiliación: Estados Unidos