Your browser doesn't support javascript.
loading
Neutrophil extracellular trap-derived enzymes oxidize high-density lipoprotein: an additional proatherogenic mechanism in systemic lupus erythematosus.
Smith, Carolyne K; Vivekanandan-Giri, Anuradha; Tang, Chongren; Knight, Jason S; Mathew, Anna; Padilla, Robin L; Gillespie, Brenda W; Carmona-Rivera, Carmelo; Liu, Xiaodan; Subramanian, Venkataraman; Hasni, Sarfaraz; Thompson, Paul R; Heinecke, Jay W; Saran, Rajiv; Pennathur, Subramaniam; Kaplan, Mariana J.
Afiliação
  • Smith CK; Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892.
  • Vivekanandan-Giri A; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
  • Tang C; Department of Medicine and Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109.
  • Knight JS; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
  • Mathew A; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
  • Padilla RL; Kidney Epidemiology and Cost Center and Biostatistics, University of Michigan, Ann Arbor, MI 48109.
  • Gillespie BW; Kidney Epidemiology and Cost Center and Biostatistics, University of Michigan, Ann Arbor, MI 48109.
  • Carmona-Rivera C; Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892.
  • Liu X; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
  • Subramanian V; Department of Chemistry, Scripps-Florida, Jupiter, FL 33458.
  • Hasni S; Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892.
  • Thompson PR; Department of Chemistry, Scripps-Florida, Jupiter, FL 33458.
  • Heinecke JW; Department of Medicine and Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109.
  • Saran R; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
  • Pennathur S; Kidney Epidemiology and Cost Center and Biostatistics, University of Michigan, Ann Arbor, MI 48109.
  • Kaplan MJ; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.
Arthritis Rheumatol ; 66(9): 2532-2544, 2014 Sep.
Article em En | MEDLINE | ID: mdl-24838349
ABSTRACT

OBJECTIVE:

Oxidative stress and oxidized high-density lipoprotein (HDL) are implicated as risk factors for cardiovascular disease (CVD) in systemic lupus erythematosus (SLE). Yet, how HDL is oxidized and rendered dysfunctional in SLE remains unclear. Neutrophil extracellular traps (NETs), the levels of which are elevated in lupus, possess oxidant-generating enzymes, including myeloperoxidase (MPO), NADPH oxidase (NOX), and nitric oxide synthase (NOS). We hypothesized that NETs mediate HDL oxidation, impairing cholesterol efflux capacity (CEC).

METHODS:

Plasma MPO levels and CEC activity were examined in controls and lupus patients, and 3-chlorotyrosine (MPO specific) and 3-nitrotyrosine (derived from reactive nitrogen species) were quantified in human HDL. Multivariable linear models were used to estimate and test differences between groups. HDL was exposed to NETs from control and lupus neutrophils in the presence or absence of MPO, NOX, NOS inhibitors, and chloroquine (CQ). Murine HDL oxidation was quantified after NET inhibition in vivo.

RESULTS:

SLE patients displayed higher MPO levels and diminished CEC compared to controls. SLE HDL had higher 3-nitrotyrosine and 3-chlorotyrosine content than control HDL, with site-specific oxidation signatures on apolipoprotein A-I. Experiments with human and murine NETs confirmed that chlorination was mediated by MPO and NOX, and nitration by NOS and NOX. Mice with lupus treated with the NET inhibitor Cl-amidine displayed significantly decreased HDL oxidation. CQ inhibited NET formation in vitro.

CONCLUSION:

Active NOS, NOX, and MPO within NETs significantly modify HDL, rendering the lipoprotein proatherogenic. Since NET formation is enhanced in SLE, these findings support a novel role for NET-derived lipoprotein oxidation in SLE-associated CVD and identify additional proatherogenic roles of neutrophils and putative protective roles of antimalarials in autoimmunity.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Estresse Oxidativo / Lipoproteínas HDL / Lúpus Eritematoso Sistêmico / Neutrófilos Tipo de estudo: Prognostic_studies / Risk_factors_studies Limite: Adult / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Arthritis Rheumatol Ano de publicação: 2014 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Estresse Oxidativo / Lipoproteínas HDL / Lúpus Eritematoso Sistêmico / Neutrófilos Tipo de estudo: Prognostic_studies / Risk_factors_studies Limite: Adult / Animals / Female / Humans / Male / Middle aged Idioma: En Revista: Arthritis Rheumatol Ano de publicação: 2014 Tipo de documento: Article