Your browser doesn't support javascript.
loading
Circulating sex steroids coregulate adipose tissue immune cell populations in healthy men.
Rubinow, Katya B; Chao, Jing H; Hagman, Derek; Kratz, Mario; Van Yserloo, Brian; Gaikwad, Nilesh W; Amory, John K; Page, Stephanie T.
Afiliação
  • Rubinow KB; Center for Research in Reproduction and Contraception, Department of Medicine, University of Washington School of Medicine, Seattle, Washington; rubinow@uw.edu.
  • Chao JH; Diabetes Institute, Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington.
  • Hagman D; Center for Research in Reproduction and Contraception, Department of Medicine, University of Washington School of Medicine, Seattle, Washington.
  • Kratz M; The Fred Hutchinson Cancer Research Center, Seattle, Washington; and.
  • Van Yserloo B; Department of Nutrition and Department of Environmental Toxicology, University of California-Davis, Davis, California.
  • Gaikwad NW; Department of Surgery, University of Washington School of Medicine, Seattle, Washington.
  • Amory JK; Department of Nutrition and Department of Environmental Toxicology, University of California-Davis, Davis, California.
  • Page ST; Center for Research in Reproduction and Contraception, Department of Medicine, University of Washington School of Medicine, Seattle, Washington.
Am J Physiol Endocrinol Metab ; 313(5): E528-E539, 2017 11 01.
Article em En | MEDLINE | ID: mdl-28698282
ABSTRACT
Male hypogonadism results in changes in body composition characterized by increases in fat mass. Resident immune cells influence energy metabolism in adipose tissue and could promote increased adiposity through paracrine effects. We hypothesized that manipulation of circulating sex steroid levels in healthy men would alter adipose tissue immune cell populations. Subjects (n = 44 men, 19-55 yr of age) received 4 wk of treatment with the gonadotropin-releasing hormone receptor antagonist acyline with daily administration of 1) placebo gel, 2) 1.25 g testosterone gel (1.62%), 3) 5 g testosterone gel, or 4) 5 g testosterone gel with an aromatase inhibitor. Subcutaneous adipose tissue biopsies were performed at baseline and end-of-treatment, and adipose tissue immune cells, gene expression, and intra-adipose estrogen levels were quantified. Change in serum total testosterone level correlated inversely with change in the number of CD3+ (ß = -0.36, P = 0.04), CD4+ (ß = -0.34, P = 0.04), and CD8+ (ß = -0.33, P = 0.05) T cells within adipose tissue. Change in serum 17ß-estradiol level correlated inversely with change in the number of adipose tissue macrophages (ATMs) (ß = -0.34, P = 0.05). A negative association also was found between change in serum testosterone and change in CD11c+ ATMs (ß = -0.41, P = 0.01). Overall, sex steroid deprivation was associated with increases in adipose tissue T cells and ATMs. No associations were found between changes in serum sex steroid levels and changes in adipose tissue gene expression. Circulating sex steroid levels may regulate adipose tissue immune cell populations. These exploratory findings highlight a possible novel mechanism that could contribute to increased metabolic risk in hypogonadal men.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Clinical_trials Limite: Adult / Humans / Male / Middle aged Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Clinical_trials Limite: Adult / Humans / Male / Middle aged Idioma: En Ano de publicação: 2017 Tipo de documento: Article