Your browser doesn't support javascript.
loading
Cholesterol Efflux Pathways Suppress Inflammasome Activation, NETosis, and Atherogenesis.
Westerterp, Marit; Fotakis, Panagiotis; Ouimet, Mireille; Bochem, Andrea E; Zhang, Hanrui; Molusky, Matthew M; Wang, Wei; Abramowicz, Sandra; la Bastide-van Gemert, Sacha; Wang, Nan; Welch, Carrie L; Reilly, Muredach P; Stroes, Erik S; Moore, Kathryn J; Tall, Alan R.
Afiliação
  • Westerterp M; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Fotakis P; Department of Pediatrics, Section of Molecular Genetics (M.W.).
  • Ouimet M; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Bochem AE; Department of Medicine, Division of Cardiology, New York University Medical Center, NY (M.O., K.J.M.).
  • Zhang H; University of Ottawa Heart Institute, and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.O.).
  • Molusky MM; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Wang W; Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (A.E.B., E.S.S.).
  • Abramowicz S; Division of Cardiology (H.Z., M.P.R.), Department of Medicine, Columbia University, New York, NY.
  • la Bastide-van Gemert S; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Wang N; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Welch CL; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Reilly MP; Department of Epidemiology (S.l.B-v.G.), University of Groningen, University Medical Center Groningen, The Netherlands.
  • Stroes ES; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Moore KJ; Division of Molecular Medicine (M.W., P.F., A.E.B., M.M.M., W.W., S.A., N.W., C.L.W., A.R.T.).
  • Tall AR; Division of Cardiology (H.Z., M.P.R.), Department of Medicine, Columbia University, New York, NY.
Circulation ; 138(9): 898-912, 2018 08 28.
Article em En | MEDLINE | ID: mdl-29588315
ABSTRACT

BACKGROUND:

The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) showed that antagonism of interleukin (IL)-1ß reduces coronary heart disease in patients with a previous myocardial infarction and evidence of systemic inflammation, indicating that pathways required for IL-1ß secretion increase cardiovascular risk. IL-1ß and IL-18 are produced via the NLRP3 inflammasome in myeloid cells in response to cholesterol accumulation, but mechanisms linking NLRP3 inflammasome activation to atherogenesis are unclear. The cholesterol transporters ATP binding cassette A1 and G1 (ABCA1/G1) mediate cholesterol efflux to high-density lipoprotein, and Abca1/g1 deficiency in myeloid cells leads to cholesterol accumulation.

METHODS:

To interrogate mechanisms connecting inflammasome activation with atherogenesis, we used mice with myeloid Abca1/g1 deficiency and concomitant deficiency of the inflammasome components Nlrp3 or Caspase-1/11. Bone marrow from these mice was transplanted into Ldlr-/- recipients, which were fed a Western-type diet.

RESULTS:

Myeloid Abca1/g1 deficiency increased plasma IL-18 levels in Ldlr-/- mice and induced IL-1ß and IL-18 secretion in splenocytes, which was reversed by Nlrp3 or Caspase-1/11 deficiency, indicating activation of the NLRP3 inflammasome. Nlrp3 or Caspase-1/11 deficiency decreased atherosclerotic lesion size in myeloid Abca1/g1-deficient Ldlr-/- mice. Myeloid Abca1/g1 deficiency enhanced caspase-1 cleavage not only in splenic monocytes and macrophages, but also in neutrophils, and dramatically enhanced neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques, with reversal by Nlrp3 or Caspase-1/11 deficiency, suggesting that inflammasome activation promotes neutrophil recruitment and neutrophil extracellular trap formation in atherosclerotic plaques. These effects appeared to be indirectly mediated by systemic inflammation leading to activation and accumulation of neutrophils in plaques. Myeloid Abca1/g1 deficiency also activated the noncanonical inflammasome, causing increased susceptibility to lipopolysaccharide-induced mortality. Patients with Tangier disease, who carry loss-of-function mutations in ABCA1 and have increased myeloid cholesterol content, showed a marked increase in plasma IL-1ß and IL-18 levels.

CONCLUSIONS:

Cholesterol accumulation in myeloid cells activates the NLRP3 inflammasome, which enhances neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques. Patients with Tangier disease, who have increased myeloid cholesterol content, showed markers of inflammasome activation, suggesting human relevance.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Colesterol / Células Mieloides / Aterosclerose / Inflamassomos / Transportador 1 de Cassete de Ligação de ATP / Armadilhas Extracelulares / Proteína 3 que Contém Domínio de Pirina da Família NLR / Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP / Inflamação Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Colesterol / Células Mieloides / Aterosclerose / Inflamassomos / Transportador 1 de Cassete de Ligação de ATP / Armadilhas Extracelulares / Proteína 3 que Contém Domínio de Pirina da Família NLR / Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP / Inflamação Idioma: En Ano de publicação: 2018 Tipo de documento: Article