Your browser doesn't support javascript.
loading
Microbiota-dependent signals are required to sustain TLR-mediated immune responses.
Weaver, Lehn K; Minichino, Danielle; Biswas, Chhanda; Chu, Niansheng; Lee, Jung-Jin; Bittinger, Kyle; Albeituni, Sabrin; Nichols, Kim E; Behrens, Edward M.
Afiliação
  • Weaver LK; Division of Pediatric Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • Minichino D; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
  • Biswas C; Division of Pediatric Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • Chu N; Division of Pediatric Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • Lee JJ; CHOP Microbiome Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • Bittinger K; CHOP Microbiome Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • Albeituni S; Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
  • Nichols KE; Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
  • Behrens EM; Division of Pediatric Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
JCI Insight ; 4(1)2019 Jan 10.
Article em En | MEDLINE | ID: mdl-30626747
ABSTRACT
Host-commensal interactions are critical for the generation of robust inflammatory responses, yet the mechanisms leading to this effect remain poorly understood. Using a murine model of cytokine storm, we identified that host microbiota are required to sustain systemic TLR-driven immune responses. Mice treated with broad-spectrum antibiotics or raised in germ-free conditions responded normally to an initial TLR signal but failed to sustain production of proinflammatory cytokines following administration of repeated TLR signals in vivo. Mechanistically, host microbiota primed JAK signaling in myeloid progenitors to promote TLR-enhanced myelopoiesis, which is required for the accumulation of TLR-responsive monocytes. In the absence of TLR-enhanced monocytopoiesis, antibiotic-treated mice lost their ability to respond to repeated TLR stimuli and were protected from cytokine storm-induced immunopathology. These data reveal priming of TLR-enhanced myelopoiesis as a microbiota-dependent mechanism that regulates systemic inflammatory responses and highlight a role for host commensals in the pathogenesis of cytokine storm syndromes.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article