Your browser doesn't support javascript.
loading
Chronic Adrenergic Stress Contributes to Metabolic Dysfunction and an Exhausted Phenotype in T Cells in the Tumor Microenvironment.
Qiao, Guanxi; Chen, Minhui; Mohammadpour, Hemn; MacDonald, Cameron R; Bucsek, Mark J; Hylander, Bonnie L; Barbi, Joseph J; Repasky, Elizabeth A.
Afiliação
  • Qiao G; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Chen M; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Mohammadpour H; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • MacDonald CR; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Bucsek MJ; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Hylander BL; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Barbi JJ; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Repasky EA; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York. Elizabeth.Repasky@RoswellPark.org.
Cancer Immunol Res ; 9(6): 651-664, 2021 06.
Article em En | MEDLINE | ID: mdl-33762351
ABSTRACT
Metabolic dysfunction and exhaustion in tumor-infiltrating T cells have been linked to ineffectual antitumor immunity and the failure of immune checkpoint inhibitor therapy. We report here that chronic stress plays a previously unrecognized role in regulating the state of T cells in the tumor microenvironment (TME). Using two mouse tumor models, we found that blocking chronic adrenergic stress signaling using the pan ß-blocker propranolol or by using mice lacking the ß2-adrenergic receptor (ß2-AR) results in reduced tumor growth rates with significantly fewer tumor-infiltrating T cells that express markers of exhaustion, with a concomitant increase in progenitor exhausted T cells. We also report that blocking ß-AR signaling in mice increases glycolysis and oxidative phosphorylation in tumor-infiltrating lymphocytes (TIL), which associated with increased expression of the costimulatory molecule CD28 and increased antitumor effector functions, including increased cytokine production. Using T cells from Nur77-GFP reporter mice to monitor T-cell activation, we observed that stress-induced ß-AR signaling suppresses T-cell receptor (TCR) signaling. Together, these data suggest that chronic stress-induced adrenergic receptor signaling serves as a "checkpoint" of immune responses and contributes to immunosuppression in the TME by promoting T-cell metabolic dysfunction and exhaustion. These results also support the possibility that chronic stress, which unfortunately is increased in many patients with cancer following their diagnoses, could be exerting a major negative influence on the outcome of therapies that depend upon the status of TILs and support the use of strategies to reduce stress or ß-AR signaling in combination with immunotherapy.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos do Interstício Tumoral / Receptores Adrenérgicos beta 2 / Linfócitos T CD8-Positivos / Microambiente Tumoral / Neoplasias Experimentais Limite: Animals Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos do Interstício Tumoral / Receptores Adrenérgicos beta 2 / Linfócitos T CD8-Positivos / Microambiente Tumoral / Neoplasias Experimentais Limite: Animals Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2021 Tipo de documento: Article
...