Your browser doesn't support javascript.
loading
Semaphorin 3F signaling actively retains neutrophils at sites of inflammation.
Plant, Tracie; Eamsamarng, Suttida; Sanchez-Garcia, Manuel A; Reyes, Leila; Renshaw, Stephen A; Coelho, Patricia; Mirchandani, Ananda S; Morgan, Jessie-May; Ellett, Felix E; Morrison, Tyler; Humphries, Duncan; Watts, Emily R; Murphy, Fiona; Raffo-Iraolagoitia, Ximena L; Zhang, Ailiang; Cash, Jenna L; Loynes, Catherine; Elks, Philip M; Van Eeden, Freek; Carlin, Leo M; Furley, Andrew Jw; Whyte, Moira Kb; Walmsley, Sarah R.
Afiliação
  • Plant T; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Eamsamarng S; Department of Infection, Immunity and Cardiovascular Disease and.
  • Sanchez-Garcia MA; Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
  • Reyes L; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Renshaw SA; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Coelho P; Department of Infection, Immunity and Cardiovascular Disease and.
  • Mirchandani AS; Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
  • Morgan JM; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Ellett FE; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Morrison T; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Humphries D; Department of Infection, Immunity and Cardiovascular Disease and.
  • Watts ER; Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
  • Murphy F; BioMEMS Resource Centre, Division of Surgery, Innovation and Bioengineering, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Raffo-Iraolagoitia XL; Burn Care, Shriners Hospitals for Children - Boston, Boston, Massachusetts, USA.
  • Zhang A; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Cash JL; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Loynes C; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Elks PM; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Van Eeden F; Cancer Research UK Beatson Institute, Glasgow, United Kingdom.
  • Carlin LM; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Furley AJ; University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
  • Whyte MK; Department of Infection, Immunity and Cardiovascular Disease and.
  • Walmsley SR; Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
J Clin Invest ; 130(6): 3221-3237, 2020 06 01.
Article em En | MEDLINE | ID: mdl-32191647
ABSTRACT
Neutrophilic inflammation is central to disease pathogenesis, for example, in chronic obstructive pulmonary disease, yet the mechanisms that retain neutrophils within tissues remain poorly understood. With emerging evidence that axon guidance factors can regulate myeloid recruitment and that neutrophils can regulate expression of a class 3 semaphorin, SEMA3F, we investigated the role of SEMA3F in inflammatory cell retention within inflamed tissues. We observed that neutrophils upregulate SEMA3F in response to proinflammatory mediators and following neutrophil recruitment to the inflamed lung. In both zebrafish tail injury and murine acute lung injury models of neutrophilic inflammation, overexpression of SEMA3F delayed inflammation resolution with slower neutrophil migratory speeds and retention of neutrophils within the tissues. Conversely, constitutive loss of sema3f accelerated egress of neutrophils from the tail injury site in fish, whereas neutrophil-specific deletion of Sema3f in mice resulted in more rapid neutrophil transit through the airways, and significantly reduced time to resolution of the neutrophilic response. Study of filamentous-actin (F-actin) subsequently showed that SEMA3F-mediated retention is associated with F-actin disassembly. In conclusion, SEMA3F signaling actively regulates neutrophil retention within the injured tissues with consequences for neutrophil clearance and inflammation resolution.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Peixe-Zebra / Transdução de Sinais / Movimento Celular / Proteínas de Peixe-Zebra / Proteínas de Membrana / Proteínas do Tecido Nervoso / Neutrófilos Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Reino Unido

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Peixe-Zebra / Transdução de Sinais / Movimento Celular / Proteínas de Peixe-Zebra / Proteínas de Membrana / Proteínas do Tecido Nervoso / Neutrófilos Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Reino Unido