Your browser doesn't support javascript.
loading
Leptin directly promotes T-cell glycolytic metabolism to drive effector T-cell differentiation in a mouse model of autoimmunity.
Gerriets, Valerie A; Danzaki, Keiko; Kishton, Rigel J; Eisner, William; Nichols, Amanda G; Saucillo, Donte C; Shinohara, Mari L; MacIver, Nancie J.
Affiliation
  • Gerriets VA; Department of Pediatrics, Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC, USA.
  • Danzaki K; Department of Pediatrics, Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC, USA.
  • Kishton RJ; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
  • Eisner W; Department of Pediatrics, Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC, USA.
  • Nichols AG; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
  • Saucillo DC; Department of Pediatrics, Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC, USA.
  • Shinohara ML; Department of Immunology, Duke University Medical Center, Durham, NC, USA.
  • MacIver NJ; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA.
Eur J Immunol ; 46(8): 1970-83, 2016 08.
Article in En | MEDLINE | ID: mdl-27222115
ABSTRACT
Upon activation, T cells require energy for growth, proliferation, and function. Effector T (Teff) cells, such as Th1 and Th17 cells, utilize high levels of glycolytic metabolism to fuel proliferation and function. In contrast, Treg cells require oxidative metabolism to fuel suppressive function. It remains unknown how Teff/Treg-cell metabolism is altered when nutrients are limited and leptin levels are low. We therefore examined the role of malnutrition and associated hypoleptinemia on Teff versus Treg cells. We found that both malnutrition-associated hypoleptinemia and T cell-specific leptin receptor knockout suppressed Teff-cell number, function, and glucose metabolism, but did not alter Treg-cell metabolism or suppressive function. Using the autoimmune mouse model EAE, we confirmed that fasting-induced hypoleptinemia altered Teff-cell, but not Treg-cell, glucose metabolism, and function in vivo, leading to decreased disease severity. To explore potential mechanisms, we examined HIF-1α, a key regulator of Th17 differentiation and Teff-cell glucose metabolism, and found HIF-1α expression was decreased in T cell-specific leptin receptor knockout Th17 cells, and in Teff cells from fasted EAE mice, but was unchanged in Treg cells. Altogether, these data demonstrate a selective, cell-intrinsic requirement for leptin to upregulate glucose metabolism and maintain function in Teff, but not Treg cells.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: T-Lymphocytes, Regulatory / Leptin / Malnutrition / Encephalomyelitis, Autoimmune, Experimental / Th17 Cells Type of study: Prognostic_studies Limits: Animals Language: En Journal: Eur J Immunol Year: 2016 Type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: T-Lymphocytes, Regulatory / Leptin / Malnutrition / Encephalomyelitis, Autoimmune, Experimental / Th17 Cells Type of study: Prognostic_studies Limits: Animals Language: En Journal: Eur J Immunol Year: 2016 Type: Article Affiliation country: United States