There has been a striking rise in obesity over the last two decades, together with parallel increases in incidences of insulin resistance, type-2 diabetes and cardiovascular disease. A critical link between obesity and downstream cardiometabolic disorders is chronic, low-grade inflammation, promoted by secretion of pro- inflammatory mediators by adipocytes and macrophages in visceral adipose tissue (VAT). While several positive effectors of adipose-tissue inflammation have been identified and characterized, negative regulators remain poorly defined. In 2009, we reported a population of Foxp3+CD4+ regulatory T (Treg) cells - with a distinct gene-expression profile and T cell receptor repertoire that is highly enriched in the VAT, but not lymphoid tissue, of lean mice as they age. This population is strikingly and specifically reduced in several insulin-resistant mouse models of obesity. Loss- and gain-of-function experiments demonstrated that Tregs can indeed regulate adipose tissue inflammation, as well as local and systemic metabolic indices. VAT Tregs have proven to be a paradigm for regulatory T cell populations specifically adapted to survive and function at particular tissue locales. Over the last funding cycle, we made substantial strides in elucidating the biology of VAT Tregs: identifying their defining transcription factor, PPAR?; extending the evidence for their role in controlling metabolic indices; delineating factors that drive their accumulation in aging lean mice, notably Ag/MHCII and the IL-1 family member, IL-33; dissecting their response to obesity; and transposing the tissue Treg concept to skeletal muscle. Building on these findings, the overall goal of this proposed project is to elucidate the molecular pathways that control VAT Treg dynamics.
Three Specific Aims will undertaken, each designed to address a hypothesis that emerged from results obtained during the last funding cycle.
The Aims are connected by their potential to explicate the generation of specialized tissular Treg populations. We propose to: 1) Elucidate how IL-33 drives accumulation of Tregs in VAT. 2) Determine whether and how TNF-a drives the loss of VAT Tregs in obese mice. 3) Determine how PPAR? and Foxp3 collaborate to impose the VAT Treg phenotype. These studies should yield important new information on molecular pathways involved in regulating the devastating downstream consequences of obesity. Identification of novel therapeutic targets within these pathways is especially important given unacceptable side-effects of the front-line thiazolidinedione family of type-2 diabetes drugs.

Public Health Relevance

This project focuses on the molecular pathways controlling the homeostasis of a unique population of regulatory T cells that resides in the visceral adipose tissue of lean, but not obese, mice and humans, and is capable of controlling insulin resistance and, thereby, type-2 diabetes and cardiovascular diseases. Obesity and downstream cardiometabolic disorders are major health challenges of the 21st century. Specifically enhancing the representation of adipose-tissue Tregs, their activity, or certain of their effector pathways or molecules offers a potential alternative to the current front-line thiazolidinedione (TZD) drugs, whose side-effects have elicited concern of late.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK092541-08
Application #
9533182
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Abraham, Kristin M
Project Start
2011-07-12
Project End
2019-07-31
Budget Start
2018-08-01
Budget End
2019-07-31
Support Year
8
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Harvard Medical School
Department
Pathology
Type
Schools of Medicine
DUNS #
047006379
City
Boston
State
MA
Country
United States
Zip Code
Li, Chaoran; DiSpirito, Joanna R; Zemmour, David et al. (2018) TCR Transgenic Mice Reveal Stepwise, Multi-site Acquisition of the Distinctive Fat-Treg Phenotype. Cell 174:285-299.e12
DiSpirito, Joanna R; Zemmour, David; Ramanan, Deepshika et al. (2018) Molecular diversification of regulatory T cells in nonlymphoid tissues. Sci Immunol 3:
Kuswanto, Wilson; Burzyn, Dalia; Panduro, Marisella et al. (2016) Poor Repair of Skeletal Muscle in Aging Mice Reflects a Defect in Local, Interleukin-33-Dependent Accumulation of Regulatory T Cells. Immunity 44:355-67
Panduro, Marisella; Benoist, Christophe; Mathis, Diane (2016) Tissue Tregs. Annu Rev Immunol 34:609-33
Mathis, Diane (2016) IL-33, Imprimatur of Adipocyte Thermogenesis. Cell 166:794-795
DiSpirito, Joanna R; Mathis, Diane (2015) Immunological contributions to adipose tissue homeostasis. Semin Immunol 27:315-21
Kolodin, Dmitriy; van Panhuys, Nicolas; Li, Chaoran et al. (2015) Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice. Cell Metab 21:543-57
Cipolletta, Daniela; Cohen, Paul; Spiegelman, Bruce M et al. (2015) Appearance and disappearance of the mRNA signature characteristic of Treg cells in visceral adipose tissue: age, diet, and PPAR? effects. Proc Natl Acad Sci U S A 112:482-7
Cohen, Paul; Levy, Julia D; Zhang, Yingying et al. (2014) Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch. Cell 156:304-16
Burzyn, Dalia; Benoist, Christophe; Mathis, Diane (2013) Regulatory T cells in nonlymphoid tissues. Nat Immunol 14:1007-13

Showing the most recent 10 out of 17 publications