Your browser doesn't support javascript.
loading
A Limited Role for AMD3100 Induced Stem Cell Mobilization for Modulation of Thoracic Trauma Outcome.
Vogel, Mona; Möhrle, Bettina; Sakk, Vadim; Brown, Andreas; Palmer, Annette; Braumüller, Sonja; Huber-Lang, Markus; Allgöwer, Andreas; Cancelas, Jose A; Geiger, Hartmut.
Afiliação
  • Vogel M; Institute of Molecular Medicine, Ulm University, Ulm, Germany.
  • Möhrle B; Institute of Molecular Medicine, Ulm University, Ulm, Germany.
  • Sakk V; Institute of Molecular Medicine, Ulm University, Ulm, Germany.
  • Brown A; Institute of Molecular Medicine, Ulm University, Ulm, Germany.
  • Palmer A; Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany.
  • Braumüller S; Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany.
  • Huber-Lang M; Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany.
  • Allgöwer A; Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany.
  • Cancelas JA; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio.
  • Geiger H; Institute of Molecular Medicine, Ulm University, Ulm, Germany.
Shock ; 57(6): 260-267, 2022 06 01.
Article em En | MEDLINE | ID: mdl-35759306
ABSTRACT
ABSTRACT Thoracic trauma is a major cause of mortality due to the associated inflammatory acute respiratory distress syndrome and morbidity due to impaired tissue regeneration. Trauma-induced lung inflammation is characterized by the early recruitment of cells with pro- or anti-inflammatory activity to the lung. Therapeutic interventions reducing the level of tissue inflammation may result in decreased tissue damage and improved healing and recovery. Stem cells might be able to improve trauma outcome via immunomodulation or by enhancing tissue regeneration.Here, we describe the migratory dynamics of murine mesenchymal, hematopoietic and endothelial stem and progenitor cells (SPCs) as well as mature inflammatory cells (monocytes, neutrophils, lymphocytes) to peripheral blood (PB) and lung tissue between 0.2 and 48 h post-blunt chest trauma (TXT). We demonstrate that the kinetics of immune cell and SPC distribution upon trauma are both cell-type and tissue-dependent. We identified a transient, early increase in the number of inflammatory cells in PB and lung at 2 h post-TXT and a second wave of infiltrating SPCs in lungs by 48 h after TXT induction, suggesting a role for SPCs in tissue remodeling after the initial inflammatory phase. Cxcl12/Cxcr4 blockade by AMD3100 within the first 6 h after TXT, while inducing a strong and coordinated mobilization of SPCs and leukocytes to PB and lung tissue, did not significantly affect TXT associated inflammation or tissue damage as determined by inflammatory cytokine levels, plasma markers for organ function, lung cell proliferation and survival, and myofibroblast/fibroblast ratio in the lung. Further understanding the dynamics of the distribution of endogenous SPCs and inflammatory cells will therefore be indispensable for stem cell-based or immunomodulation therapies in trauma.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Traumatismos Torácicos / Ferimentos não Penetrantes Limite: Animals Idioma: En Revista: Shock Assunto da revista: ANGIOLOGIA / CARDIOLOGIA Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Traumatismos Torácicos / Ferimentos não Penetrantes Limite: Animals Idioma: En Revista: Shock Assunto da revista: ANGIOLOGIA / CARDIOLOGIA Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Alemanha