Your browser doesn't support javascript.
loading
Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert.
Lee, Geoffrey; Espirito Santo, Ana Isabel; Zwingenberger, Stefan; Cai, Lawrence; Vogl, Thomas; Feldmann, Marc; Horwood, Nicole J; Chan, James K; Nanchahal, Jagdeep.
Afiliação
  • Lee G; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom.
  • Espirito Santo AI; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom.
  • Zwingenberger S; University Center of Orthopaedics and Traumatology, University Hospital Carl Gustav Carus at Technische Universität Dresden, 01307 Dresden, Germany.
  • Cai L; Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
  • Vogl T; Institute of Immunology, University of Münster, 48149 Münster, Germany.
  • Feldmann M; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom; marc.feldmann@kennedy.ox.ac.uk jagdeep.nanchahal@kennedy.ox.ac.uk.
  • Horwood NJ; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom.
  • Chan JK; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom.
  • Nanchahal J; Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, OX3 7FY Oxford, United Kingdom; marc.feldmann@kennedy.ox.ac.uk jagdeep.nanchahal@kennedy.ox.ac.uk.
Proc Natl Acad Sci U S A ; 115(19): E4463-E4472, 2018 05 08.
Article em En | MEDLINE | ID: mdl-29674451
ABSTRACT
A major discovery of recent decades has been the existence of stem cells and their potential to repair many, if not most, tissues. With the aging population, many attempts have been made to use exogenous stem cells to promote tissue repair, so far with limited success. An alternative approach, which may be more effective and far less costly, is to promote tissue regeneration by targeting endogenous stem cells. However, ways of enhancing endogenous stem cell function remain poorly defined. Injury leads to the release of danger signals which are known to modulate the immune response, but their role in stem cell-mediated repair in vivo remains to be clarified. Here we show that high mobility group box 1 (HMGB1) is released following fracture in both humans and mice, forms a heterocomplex with CXCL12, and acts via CXCR4 to accelerate skeletal, hematopoietic, and muscle regeneration in vivo. Pretreatment with HMGB1 2 wk before injury also accelerated tissue regeneration, indicating an acquired proregenerative signature. HMGB1 led to sustained increase in cell cycling in vivo, and using Hmgb1-/- mice we identified the underlying mechanism as the transition of multiple quiescent stem cells from G0 to GAlert HMGB1 also transitions human stem and progenitor cells to GAlert Therefore, exogenous HMGB1 may benefit patients in many clinical scenarios, including trauma, chemotherapy, and elective surgery.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regeneração / Células-Tronco Hematopoéticas / Ciclo Celular / Músculo Esquelético / Proteína HMGB1 / Fraturas Ósseas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regeneração / Células-Tronco Hematopoéticas / Ciclo Celular / Músculo Esquelético / Proteína HMGB1 / Fraturas Ósseas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2018 Tipo de documento: Article