Your browser doesn't support javascript.
loading
Integrin α4ß1/VCAM-1 Interaction Evokes Dynamic Cell Aggregation Between Immune Cells and Human Lung Microvascular Endothelial Cells at Infectious Hemolysis.
Lou, Hai-Yan; Yan, Hai-Peng; Yang, Long-Gui; Fan, Jiang-Hua; Cho, William C; Xiao, Zheng-Hui; Li, Shuang-Jie.
Afiliación
  • Lou HY; Emergency Center of Hunan Children's Hospital, Changsha, China.
  • Yan HP; Department of Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, China.
  • Yang LG; Emergency Center of Hunan Children's Hospital, Changsha, China.
  • Fan JH; Emergency Center of Hunan Children's Hospital, Changsha, China.
  • Cho WC; Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China.
  • Xiao ZH; Emergency Center of Hunan Children's Hospital, Changsha, China.
  • Li SJ; Department of Hepatopathy, Hunan Children's Hospital, Changsha, China.
Front Pharmacol ; 12: 653143, 2021.
Article en En | MEDLINE | ID: mdl-33959020
ABSTRACT
Bacterial and viral infection is a common cause of pneumonia, respiratory failure, and even acute respiratory distress syndrome. Increasing evidence indicates that red blood cells (RBCs) may contribute to immune response and inflammation. However, the precise molecular mechanisms that link RBC and hemolysis to the development and progression of inflammatory pathologies are not entirely understood. In this study, we used bacterial endotoxin, lipopolysaccharide (LPS), to mimic an infectious hemolysis and found that RBCs dynamically regulated cell aggregation between immune cells and human lung microvascular endothelial cells (HLMVEC). When RBCs were treated with LPS, integrin α4ß1 was increased and was accompanied by cytokines and chemokines release (TNF-α, IL-1ß, IL-6, IL-8, IFN-γ, CXCL12, CCL5, CCL7 and CCL4). Upon α4ß1 elevation, RBCs not only facilitated mature monocyte derived dendritic cell (mo-DCs) adhesion but also promoted HLMVEC aggregation. Furthermore, co-culture of the supernatant of LPS pre-treated RBCs with mo-DCs could promote naïve CD4 T cell proliferation. Notably, the filtered culture from LPS-lysed RBCs further promoted mo-DCs migration in a concentration dependent manner. From a therapeutic perspective, cyclic peptide inhibitor of integrin α4ß1 combined with methylprednisolone (α4ß1/Methrol) remarkably blocked RBCs aggregation to mo-DCs, HLMVEC, or mo-DCs and HLMVEC mixture. Moreover, α4ß1/Methrol dramatically reduced mo-DCs migration up-regulated glucocorticoid-induced leucine zipper in mo-DCs, and ultimately reversed immune cell dysfunction induced by hemolysis. Taken together, these results indicate that integrin α4ß1 on RBCs could mediate cell-cell interaction for adaptive immunity through influencing cell adhesion, migration, and T cell proliferation.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Front Pharmacol Año: 2021 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Front Pharmacol Año: 2021 Tipo del documento: Article País de afiliación: China