Your browser doesn't support javascript.
loading
Stressed erythrophagocytosis induces immunosuppression during sepsis through heme-mediated STAT1 dysregulation.
Olonisakin, Tolani F; Suber, Tomeka; Gonzalez-Ferrer, Shekina; Xiong, Zeyu; Peñaloza, Hernán F; van der Geest, Rick; Xiong, Yuting; Osei-Hwedieh, David O; Tejero, Jesús; Rosengart, Matthew R; Mars, Wendy M; Van Tyne, Daria; Perlegas, Andreas; Brashears, Samuel; Kim-Shapiro, Daniel B; Gladwin, Mark T; Bachman, Michael A; Hod, Eldad A; St Croix, Claudette; Tyurina, Yulia Y; Kagan, Valerian E; Mallampalli, Rama K; Ray, Anuradha; Ray, Prabir; Lee, Janet S.
Afiliação
  • Olonisakin TF; Medical Scientist Training Program.
  • Suber T; Acute Lung Injury Center of Excellence.
  • Gonzalez-Ferrer S; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Xiong Z; Acute Lung Injury Center of Excellence.
  • Peñaloza HF; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • van der Geest R; Acute Lung Injury Center of Excellence.
  • Xiong Y; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Osei-Hwedieh DO; Acute Lung Injury Center of Excellence.
  • Tejero J; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Rosengart MR; Acute Lung Injury Center of Excellence.
  • Mars WM; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Van Tyne D; Acute Lung Injury Center of Excellence.
  • Perlegas A; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Brashears S; Acute Lung Injury Center of Excellence.
  • Kim-Shapiro DB; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Gladwin MT; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Bachman MA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
  • Hod EA; Vascular Medicine Institute.
  • St Croix C; Department of Surgery.
  • Tyurina YY; Department of Pathology, and.
  • Kagan VE; Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
  • Mallampalli RK; Department of Physics and The Translational Science Center, Wake Forest University, Winston-Salem, North Carolina, USA.
  • Ray A; Department of Physics and The Translational Science Center, Wake Forest University, Winston-Salem, North Carolina, USA.
  • Ray P; Department of Physics and The Translational Science Center, Wake Forest University, Winston-Salem, North Carolina, USA.
  • Lee JS; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.
J Clin Invest ; 131(1)2021 01 04.
Article em En | MEDLINE | ID: mdl-32941182
ABSTRACT
Macrophages are main effectors of heme metabolism, increasing transiently in the liver during heightened disposal of damaged or senescent RBCs (sRBCs). Macrophages are also essential in defense against microbial threats, but pathological states of heme excess may be immunosuppressive. Herein, we uncovered a mechanism whereby an acute rise in sRBC disposal by macrophages led to an immunosuppressive phenotype after intrapulmonary Klebsiella pneumoniae infection characterized by increased extrapulmonary bacterial proliferation and reduced survival from sepsis in mice. The impaired immunity to K. pneumoniae during heightened sRBC disposal was independent of iron acquisition by bacterial siderophores, in that K. pneumoniae mutants lacking siderophore function recapitulated the findings observed with the WT strain. Rather, sRBC disposal induced a liver transcriptomic profile notable for suppression of Stat1 and IFN-related responses during K. pneumoniae sepsis. Excess heme handling by macrophages recapitulated STAT1 suppression during infection that required synergistic NRF1 and NRF2 activation but was independent of heme oxygenase-1 induction. Whereas iron was dispensable, the porphyrin moiety of heme was sufficient to mediate suppression of STAT1-dependent responses in human and mouse macrophages and promoted liver dissemination of K. pneumoniae in vivo. Thus, cellular heme metabolism dysfunction negatively regulated the STAT1 pathway, with implications in severe infection.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fagocitose / Regulação da Expressão Gênica / Sepse / Eritrócitos / Fator de Transcrição STAT1 / Heme / Tolerância Imunológica Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fagocitose / Regulação da Expressão Gênica / Sepse / Eritrócitos / Fator de Transcrição STAT1 / Heme / Tolerância Imunológica Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2021 Tipo de documento: Article