Your browser doesn't support javascript.
loading
Activation of ß2-Adrenergic Receptor Promotes Growth and Angiogenesis in Breast Cancer by Down-regulating PPARγ.
Zhou, Jing; Liu, Zhanzhao; Zhang, Lingjing; Hu, Xiao; Wang, Zhihua; Ni, Hong; Wang, Yue; Qin, Junfang.
Afiliação
  • Zhou J; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Liu Z; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Zhang L; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Hu X; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Wang Z; Department of Pediatrics, Tianjin Nankai Hospital, Tianjin, China.
  • Ni H; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Wang Y; Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
  • Qin J; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China.
Cancer Res Treat ; 52(3): 830-847, 2020 Jul.
Article em En | MEDLINE | ID: mdl-32138468
ABSTRACT

PURPOSE:

Chronic stress and related hormones are key in cancer progression. Peroxisome proliferator-activated receptor γ (PPARγ) and its agonists was reported that inducing anti-tumor effect. However, the function of PPARγ in pro-tumorigenic effects induced by chronic stress in breast cancer remains unknown. Herein, we have characterized a novel role of PPARγ and vascular endothelial growth factor (VEGF)/fibroblast growth factor 2 (FGF2) signals in breast cancer promoted by chronic stress. MATERIALS AND

METHODS:

We performed experiments in vivo and in vitro and used bioinformatics data to evaluate the therapeutic potential of PPARγ in breast cancer promoted by stress.

RESULTS:

Chronic stress significantly inhibited the PPARγ expression and promoted breast cancer in vivo. VEGF/FGF2-mediated angiogenesis increased in the chronic stress group compared to the control group. PPARγ agonist pioglitazone (PioG) injection offset the pro-tumorigenic effect of chronic stress. Moreover, specific ß2-adrenergic receptor (ß2R) antagonist ICI11-8551 inhibited the effect of chronic stress. In vitro, norepinephrine (NE) treatment had a similar tendency to chronic stress. The effect of NE was mediated by the ß2R/adenylate cyclase signaling pathway and suppressed by PioG. PPARγ suppressed VEGF/FGF2 through reactive oxygen species inhibition. Bioinformatics data confirmed that therewas a lowPPARγ expression in breast invasive carcinoma. Lower PPARγ was associated with a significantly worse survival.

CONCLUSION:

ß2R activation induced by chronic stress and related hormones promotes growth and VEGF/FGF2-mediated angiogenesis of breast cancer by down-regulating PPARγ. Our findings hint that ß receptor and PPARγ as two target molecules and the novel role for their agonists or antagonists as clinical medicine in breast cancer therapy.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Receptores Adrenérgicos beta 2 / PPAR gama / Pioglitazona / Hipoglicemiantes / Neoplasias Pulmonares / Neovascularização Patológica Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Receptores Adrenérgicos beta 2 / PPAR gama / Pioglitazona / Hipoglicemiantes / Neoplasias Pulmonares / Neovascularização Patológica Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2020 Tipo de documento: Article