Your browser doesn't support javascript.
loading
Stress-Inducible Gene Atf3 Dictates a Dichotomous Macrophage Activity in Chemotherapy-Enhanced Lung Colonization.
Middleton, Justin D; Fehlman, Jared; Sivakumar, Subhakeertana; Stover, Daniel G; Hai, Tsonwin.
Afiliação
  • Middleton JD; Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA.
  • Fehlman J; Molecular, Cellular and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA.
  • Sivakumar S; Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA.
  • Stover DG; Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA.
  • Hai T; Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, OH 43210, USA.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article em En | MEDLINE | ID: mdl-34298975
ABSTRACT
Previously, we showed that chemotherapy paradoxically exacerbated cancer cell colonization at the secondary site in a manner dependent on Atf3, a stress-inducible gene, in the non-cancer host cells. Here, we present evidence that this phenotype is established at an early stage of colonization within days of cancer cell arrival. Using mouse breast cancer models, we showed that, in the wild-type (WT) lung, cyclophosphamide (CTX) increased the ability of the lung to retain cancer cells in the vascular bed. Although CTX did not change the WT lung to affect cancer cell extravasation or proliferation, it changed the lung macrophage to be pro-cancer, protecting cancer cells from death. This, combined with the initial increase in cell retention, resulted in higher lung colonization in CTX-treated than control-treated mice. In the Atf3 knockout (KO) lung, CTX also increased the ability of lung to retain cancer cells. However, the CTX-treated KO macrophage was highly cytotoxic to cancer cells, resulting in no increase in lung colonization-despite the initial increase in cell retention. In summary, the status of Atf3 dictates the dichotomous activity of macrophage pro-cancer for CTX-treated WT macrophage but anti-cancer for the KO counterpart. This dichotomy provides a mechanistic explanation for CTX to exacerbate lung colonization in the WT but not Atf3 KO lung.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Estresse Fisiológico / Ciclofosfamida / Fator 3 Ativador da Transcrição / Macrófagos Associados a Tumor / Neoplasias Pulmonares / Macrófagos / Neoplasias Mamárias Experimentais / Metástase Neoplásica / Proteínas de Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Estresse Fisiológico / Ciclofosfamida / Fator 3 Ativador da Transcrição / Macrófagos Associados a Tumor / Neoplasias Pulmonares / Macrófagos / Neoplasias Mamárias Experimentais / Metástase Neoplásica / Proteínas de Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article