The Impact of Long-term Physical Inactivity on Adipose Tissue Immunometabolism

William V Trim, Jean-Philippe Walhin, Francoise Koumanov, Anne Bouloumié, Mark A Lindsay, Rebecca L Travers, James E Turner, Dylan Thompson, William V Trim, Jean-Philippe Walhin, Francoise Koumanov, Anne Bouloumié, Mark A Lindsay, Rebecca L Travers, James E Turner, Dylan Thompson

Abstract

Context: Adipose tissue and physical inactivity both influence metabolic health and systemic inflammation, but how adipose tissue responds to chronic physical inactivity is unknown.

Objective: This work aimed to characterize the impact of chronic physical inactivity on adipose tissue in healthy, young males.

Methods: We collected subcutaneous adipose tissue from 20 healthy, young men before and after 60 days of complete bed rest with energy intake reduced to maintain energy balance and fat mass. We used RNA sequencing, flow cytometry, ex vivo tissue culture, and targeted protein analyses to examine adipose tissue phenotype.

Results: Our results indicate that the adipose tissue transcriptome, stromal cellular compartment, and insulin signaling protein abundance are largely unaffected by bed rest when fat mass is kept stable. However, there was an increase in the circulating concentration of several adipokines, including plasma leptin, which was associated with inactivity-induced increases in plasma insulin and absent from adipose tissue cultured ex vivo under standardized culture conditions.

Conclusion: Physical inactivity-induced disturbances to adipokine concentrations such as leptin, without changes to fat mass, could have profound metabolic implications outside a clinical facility when energy intake is not tightly controlled.

Trial registration: ClinicalTrials.gov NCT03594799.

Keywords: Adipose tissue; immunometabolism; physical inactivity.

© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.

Figures

Figure 1.
Figure 1.
Overview of adipose tissue transcriptional responses to long-term bed rest. (A) Volcano plot of all transcripts by Log2FC against DESeq2-produced p-values. Horizontal dotted lines indicate significance threshold (P < .01). Vertical dotted lines represent 0.5 Log2 fold change thresholds. Red dots = significantly downregulated; blue dots = significantly upregulated; grey dots = non-differentially expressed genes. (B) Quantification of differentially expressed genes identified in (A). (C) Top-10 significantly down/upregulated transcripts, by magnitude of Log2FC (presented as mean ± SEM). (D) Top 25 most significantly DEGs ranked by significance, from left to right (presented as mean ± SEM). (E) Significantly enriched (P < .05; EASE score) pathways associated with DESeq2 DEGs from (A) using DAVID bioinformatics resources 6.8. P values are presented as –Log10 values. Pathways and terms are ranked in ascending order by significance (EASE score). The specific number of differentially expressed genes within a given pathway related to differentially expressed genes put into the analyses are presented within each respective bar. A minimum threshold of 2 genes within a given pathway/term was set for consideration. BP, biological process; CC, cellular compartment; DAVID, database for annotation, visualization, and integrated discovery; DEG, differentially expressed gene; EASE, expression analysis systematic explorer; GO, gene ontology; KEGG, Kyoto encyclopedia of genes and genomes; MF, molecular function.
Figure 2.
Figure 2.
Plasma glucose and insulin, and metabolic gene and protein expression in adipose tissue in response to bed rest. (A) Fasting plasma insulin concentrations before and after bed rest. Data represent group means at each time point with individual responses overlaid. (B) Fasting plasma glucose concentrations before and after bed rest. Data represent group means at each time point with individual responses overlaid. (C) Relative gene expression of several genes associated with adipose tissue metabolism at post bed rest relative to pre bed rest. Ratios represent the fold change at post bed rest compared with pre bed rest expression levels. Data were normalized to peptidylprolyl isomerase A, pre bed rest, and internal calibrator using the ∆∆Ct method. Dashed line represents no change relative to pre bed rest. Data are presented as mean ± SEM, with the sample size indicated at the bottom of each bar. (D, E) Changes in protein expression of key insulin signaling proteins in (D) adipose tissue and (E) paired adipocytes as a ratio of post bed rest compared with pre bed rest levels, with representative blots (right of respective figure). Solid lines represent mean ± SEM, with individual responses overlaid. Dashed lines in Panel C represent no change relative to pre bed rest. Sample sizes are indicated underneath each variable in A, B, D, and E, and within bars in C.
Figure 3.
Figure 3.
Relative change in plasma and adipose tissue supernatant concentrations of inflammatory and metabolic proteins in response to bed rest. (A) Peripheral blood plasma protein concentration expressed as the average change, calculated from each individual’s change from pre to post bed rest. Data are presented as mean ± SEM. (B) Three-hour ex vivo culture of adipose tissue explant protein secretion expressed as the average change, calculated from each individual’s change from pre to post bed rest. Data are presented as mean ± SEM. Sample sizes are indicated at the base of each respective bar. Where concentrations fell below detectable limits for the assay, n.d. is written. *P < .05. CCL, chemokine (C-C motif) ligand; CXCL, chemokine (C-X-C motif) ligand; FGF, fibroblast growth factor; GM-CSF, granulocyte-macrophage colony stimulating factor; ICAM, intracellular adhesion molecule; IFN, interferon; IL, interleukin; IP, interferon gamma inducible protein; MCP, monocyte chemotactic protein; MIP, monocyte inflammatory protein; RANTES, regulated on activation, normal T-cell expressed and secreted; SAA, serum amyloid-A; TNF, tumor necrosis factor; VCAM, vascular cell adhesion molecule; VEGF, vascular endothelial growth factory.
Figure 4.
Figure 4.
Adipose tissue and peripheral blood leukocyte responses to bed rest. (A) Adipose tissue CD4+ and CD8+ T-cells expressed as number of cells per gram of tissue. (B) Adipose tissue CD206+ macrophages and NK cells expressed as number of cells per gram of tissue. (C) Adipose tissue CD206+ macrophage HLA-DR surface expression (MFI). (D) Adipose tissue CD4+ T-cell subpopulations expressed as number of cells per gram of tissue. (E) Adipose tissue CD8+ T-cell subpopulations expressed as number of cells per gram of tissue. (F) Adipose tissue endothelial cells expressed as number of cells per gram of tissue. (G) Adipose tissue progenitor cells expressed as number of cells per gram of tissue. (H) Adipose tissue MSCA-1+ progenitor cells expressed as number of cells per gram of tissue. (I) Adipose tissue MSCA-1+ progenitor cell MSCA-1+ surface expression (MFI). (J) Peripheral blood CD4+ and CD8+ T-cells expressed as cells per μL. (K) Peripheral blood monocytes subpopulations expressed as cells per μL. (L) Peripheral blood CD4+ T-cell subpopulations expressed as cells per μL. (M) Peripheral blood CD8+ T-cell subpopulations expressed as cells per μL. All data are presented as group means within individual responses overlaid. T-cell subsets presented in (D), (E), (J), and (K) represent CD45RA+CD27+ (NA) cells; CD45RA−CD27+ (CM) cells; CD45RA−CD27− (EM) cells; CD45RA+CD27− (EMRA) cells. Adipose tissue progenitor cells represent CD45−CD31−CD34+ cells. Adipose tissue endothelial cells represent CD45−CD31+CD34+ cells. Sample sizes are indicated underneath each variable. CD, cluster of differentiation; CM, central memory; EM, effector memory; EMRA, effector memory re-expressing CD45RA; NA, naïve; PBMC, peripheral blood mononuclear cell; MFI, median fluorescence intensity; MSCA–1, mesenchymal stem cell antigen-1.
Figure 5.
Figure 5.
CD4+ T–Cell, CD8+ T–cell, and monocyte subpopulation HLA-DR expression in peripheral blood in response to bed rest. (A-D) CD4+ T-cell subpopulation HLA-DR surface expression for (A) naïve; (B) central memory; (C) effector memory; and (D) terminally differentiated effector memory cells. (E–H) CD8+ T-cell subpopulation HLA-DR surface expression for (E) naïve); (F) central memory; (G) effector memory; and (H) terminally differentiated effector memory cells. (I–K) Monocyte subpopulation HLA-DR surface expression for (I) nonclassical; (J) classical; and (K) intermediate monocytes. T-cell subsets presented in (A-H) represent CD45RA+CD27+ (NA) cells; CD45RA−CD27+ (CM) cells; CD45RA−CD27− (EM) cells; CD45RA+CD27− (EMRA) cells. Data represent HLA-DR cell surface expression on a per-cell basis. Data are presented as group means with individual responses overlaid. Sample sizes are indicated underneath each variable. CD, cluster of differentiation; CM, central memory; EM, effector memory; EMRA, effector memory re-expressing CD45RA; HLA–DR, human leukocyte antigen–DR isotype; MFI, median fluorescence intensity; NA, naïve.

References

    1. Trim W, Turner JE, Thompson D. Parallels in immunometabolic adipose tissue dysfunction with ageing and obesity. Front Immunol. 2018;9:169.
    1. Galic S, Oakhill JS, Steinberg GR. Adipose tissue as an endocrine organ. Mol Cell Endocrinol. 2010;316(2):129-139.
    1. Trayhurn P, Drevon CA, Eckel J. Secreted proteins from adipose tissue and skeletal muscle – adipokines, myokines and adipose/muscle cross-talk. Arch Physiol Biochem. 2011;117(2):47-56.
    1. Mohamed-Ali V, Goodrick S, Rawesh A, et al. . Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab. 1997;82(12):4196-4200.
    1. Dietze-Schroeder D, Sell H, Uhlig M, Koenen M, Eckel J. Autocrine action of adiponectin on human fat cells prevents the release of insulin resistance-inducing factors. Diabetes. 2005;54(7):2003-2011.
    1. Rosenbaum M, Goldsmith R, Bloomfield D, et al. . Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest. 2005;115(12):3579-3586.
    1. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542(7640):177-185.
    1. LeBlanc AD, Schneider VS, Evans HJ, Pientok C, Rowe R, Spector E. Regional changes in muscle mass following 17 weeks of bed rest. J Appl Physiol (1985). 1992;73(5):2172-2178.
    1. LeBlanc A, Schneider V, Shackelford L, et al. . Bone mineral and lean tissue loss after long duration space flight. J Musculoskelet Neuronal Interact. 2000;1(2):157-160.
    1. Sonne MP, Alibegovic AC, Højbjerre L, Vaag A, Stallknecht B, Dela F. Effect of 10 days of bedrest on metabolic and vascular insulin action: a study in individuals at risk for type 2 diabetes. J Appl Physiol (1985). 2010;108(4):830-837.
    1. Mikines KJ, Richter EA, Dela F, Galbo H. Seven days of bed rest decrease insulin action on glucose uptake in leg and whole body. J Appl Physiol (1985). 1991;70(3):1245-1254.
    1. Dirks ML, Wall BT, van de Valk B, et al. . One week of bed rest leads to substantial muscle atrophy and induces whole-body insulin resistance in the absence of skeletal muscle lipid accumulation. Diabetes. 2016;65(10):2862-2875.
    1. Trappe TA, Burd NA, Louis ES, Lee GA, Trappe SW. Influence of concurrent exercise or nutrition countermeasures on thigh and calf muscle size and function during 60 days of bed rest in women. Acta Physiol (Oxf). 2007;191(2):147-159.
    1. Schmitt DA, Schaffar L, Taylor GR, et al. . Use of bed rest and head-down tilt to simulate spaceflight-induce immune system changes. J Interferon Cytokine Res. 1996;16(2):151-157.
    1. Schmitt DA, Schwarzenberg M, Tkaczuk J, et al. . Head-down tilt bed rest and immune responses. Pflugers Arch. 2000;441(2-3 Suppl):R79-R84.
    1. Højbjerre L, Sonne MP, Alibegovic AC, et al. . Impact of physical inactivity on subcutaneous adipose tissue metabolism in healthy young male offspring of patients with type 2 diabetes. Diabetes. 2010;59(11):2790-2798.
    1. Højbjerre L, Alibegovic AC, Sonne MP, et al. . Increased lipolysis but diminished gene expression of lipases in subcutaneous adipose tissue of healthy young males with intrauterine growth retardation. J Appl Physiol (1985). 2011;111(6):1863-1870.
    1. Højbjerre L, Sonne MP, Alibegovic AC, et al. . Impact of physical inactivity on adipose tissue low-grade inflammation in first-degree relatives of type 2 diabetic patients. Diabetes Care. 2011;34(10):2265-2272.
    1. Guerra B, Ponce-González JG, Morales-Alamo D, et al. . Leptin signaling in skeletal muscle after bed rest in healthy humans. Eur J Appl Physiol. 2014;114(2):345-357.
    1. Yang C, Chen J, Wu F, et al. . Effects of 60-day head-down bed rest on osteocalcin, glycolipid metabolism and their association with or without resistance training. Clin Endocrinol (Oxf). 2014;81(5):671-678.
    1. Jurdana M, Jenko-Pražnikar Z, Mohorko N, et al. . Impact of 14-day bed rest on serum adipokines and low-grade inflammation in younger and older adults. Age (Dordr). 2015;37(6):116.
    1. Heer M, Liphardt A, Frings-Meuthen P.. Standardisation of bed rest study conditions. Vol. 1.5. In: Medicine IoA. Institute of Aerospace Medicine; 2009.
    1. Trim W, Walhin J-P, Koumanov F, et al. . Data for: The impact of long-term physical inactivity on adipose tissue immunometabolism. Figshare. Deposited August 11, 2021. Doi: 10.6084/m9.figshare.15138231
    1. Damiot A, Demangel R, Noone J, et al. . A nutrient cocktail prevents lipid metabolism alterations induced by 20 days of daily steps reduction and fructose overfeeding: result from a randomized study. J Appl Physiol (1985). 2019;126(1):88-101.
    1. Arc-Chagnaud C, Py G, Fovet T, et al. . Evaluation of an antioxidant and anti-inflammatory cocktail against human hypoactivity-induced skeletal muscle deconditioning. Front Physiol. 2020;11:71.
    1. Kelly TL, Wilson KE, Heymsfield SB. Dual energy X-ray absorptiometry body composition reference values from NHANES. PLoS One. 2009;4(9):e7038.
    1. Paradisi G, Smith L, Burtner C, et al. . Dual energy X-ray absorptiometry assessment of fat mass distribution and its association with the insulin resistance syndrome. Diabetes Care. 1999;22(8):1310-1317.
    1. Travers RL, Motta AC, Betts JA, Bouloumié A, Thompson D. The impact of adiposity on adipose tissue-resident lymphocyte activation in humans. Int J Obes (Lond). 2015;39(5):762-769.
    1. Travers RL, Motta AC, Betts JA, Thompson D. Adipose tissue metabolic and inflammatory responses to a mixed meal in lean, overweight and obese men. Eur J Nutr. 2017;56(1):375-385.
    1. Trim WV, Walhin J-P, Koumanov F, et al. . Divergent immunometabolic changes in adipose tissue and skeletal muscle with ageing in healthy humans. J Physiol. 2021.
    1. Neville MJ, Collins JM, Gloyn AL, McCarthy MI, Karpe F. Comprehensive human adipose tissue mRNA and microRNA endogenous control selection for quantitative real-time-PCR normalization. Obesity (Silver Spring). 2011;19(4):888-892.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25(4):402-408.
    1. Lamble S, Batty E, Attar M, et al. . Improved workflows for high throughput library preparation using the transposome-based Nextera system. BMC Biotechnol. 2013;13:104.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44-57.
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1-13.
    1. Sturn A, Quackenbush J, Trajanoski Z. Genesis: cluster analysis of microarray data. Bioinformatics. 2002;18(1):207-208.
    1. Hosack DA, Dennis G Jr, Sherman BT, Lane HC, Lempicki RA. Identifying biological themes within lists of genes with EASE. Genome Biol. 2003;4(10):R70.
    1. Walhin JP, Richardson JD, Betts JA, Thompson D. Exercise counteracts the effects of short-term overfeeding and reduced physical activity independent of energy imbalance in healthy young men. J Physiol. 2013;591(24):6231-6243.
    1. Holman GD, Kozka IJ, Clark AE, et al. . Cell surface labeling of glucose transporter isoform GLUT4 by bis-mannose photolabel. Correlation with stimulation of glucose transport in rat adipose cells by insulin and phorbol ester. J Biol Chem. 1990;265(30):18172-18179.
    1. Kramer A, Kümmel J, Mulder E, Gollhofer A, Frings-Meuthen P, Gruber M. High-intensity jump training is tolerated during 60 days of bed rest and is very effective in preserving leg power and lean body mass: an overview of the Cologne RSL study. PLoS One. 2017;12(1):e0169793.
    1. Bergouignan A, Momken I, Schoeller DA, et al. . Regulation of energy balance during long-term physical inactivity induced by bed rest with and without exercise training. J Clin Endocrinol Metab. 2010;95(3):1045-1053.
    1. Rudwill F, Bergouignan A, Gastebois C, et al. . Effect of enforced physical inactivity induced by 60-day of bed rest on hepatic markers of NAFLD in healthy normal-weight women. Liver Int. 2015;35(6):1700-1706.
    1. Boucher J, Masri B, Daviaud D, et al. . Apelin, a newly identified adipokine up-regulated by insulin and obesity. Endocrinology. 2005;146(4):1764-1771.
    1. Shimobayashi M, Albert V, Woelnerhanssen B, et al. . Insulin resistance causes inflammation in adipose tissue. J Clin Invest. 2018;128(4):1538-1550.
    1. Hoff P, Belavý DL, Huscher D, et al. . Effects of 60-day bed rest with and without exercise on cellular and humoral immunological parameters. Cell Mol Immunol. 2015;12(4):483-492.
    1. Ribeiro F, Alves AJ, Teixeira M, et al. . Exercise training increases interleukin-10 after an acute myocardial infarction: a randomised clinical trial. Int J Sports Med. 2012;33(3):192-198.
    1. Dorneles GP, Haddad DO, Fagundes VO, et al. . High intensity interval exercise decreases IL-8 and enhances the immunomodulatory cytokine interleukin-10 in lean and overweight-obese individuals. Cytokine. 2016;77:1-9.
    1. Shearer WT, Ochs HD, Lee BN, et al. . Immune responses in adult female volunteers during the bed-rest model of spaceflight: antibodies and cytokines. J Allergy Clin Immunol. 2009;123(4):900-905.
    1. Ostlund RE Jr, Yang JW, Klein S, Gingerich R. Relation between plasma leptin concentration and body fat, gender, diet, age, and metabolic covariates. J Clin Endocrinol Metab. 1996;81(11):3909-3913.
    1. Minokoshi Y, Toda C, Okamoto S. Regulatory role of leptin in glucose and lipid metabolism in skeletal muscle. Indian J Endocrinol Metab. 2012;16(Suppl 3):S562-S568.
    1. Ceddia RB. Direct metabolic regulation in skeletal muscle and fat tissue by leptin: implications for glucose and fatty acids homeostasis. Int J Obes (Lond). 2005;29(10):1175-1183.
    1. Stern JH, Rutkowski JM, Scherer PE. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 2016;23(5):770-784.
    1. Clément K, Biebermann H, Farooqi IS, et al. . MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med. 2018;24(5):551-555.
    1. Farooqi IS, Jebb SA, Langmack G, et al. . Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med. 1999;341(12):879-884.
    1. Farooqi IS, O’Rahilly S. 20 years of leptin: human disorders of leptin action. J Endocrinol. 2014;223(1):T63-T70.
    1. Lee MJ, Wang Y, Ricci MR, Sullivan S, Russell CD, Fried SK. Acute and chronic regulation of leptin synthesis, storage, and secretion by insulin and dexamethasone in human adipose tissue. Am J Physiol Endocrinol Metab. 2007;292(3):E858-E864.
    1. Bradley RL, Cheatham B. Regulation of ob gene expression and leptin secretion by insulin and dexamethasone in rat adipocytes. Diabetes. 1999;48(2):272-278.
    1. Barr VA, Malide D, Zarnowski MJ, Taylor SI, Cushman SW. Insulin stimulates both leptin secretion and production by rat white adipose tissue. Endocrinology. 1997;138(10):4463-4472.
    1. Fischer S, Hanefeld M, Haffner SM, et al. . Insulin-resistant patients with type 2 diabetes mellitus have higher serum leptin levels independently of body fat mass. Acta Diabetol. 2002;39(3):105-110.
    1. Gonzalez JT, Betts JA, Thompson D. Carbohydrate availability as a regulator of energy balance with exercise. Exerc Sport Sci Rev. 2019;47(4):215-222.
    1. Mayer J, Roy P, Mitra KP. Relation between caloric intake, body weight, and physical work: studies in an industrial male population in West Bengal. Am J Clin Nutr. 1956;4(2):169-175.
    1. Klein S, Coppack SW, Mohamed-Ali V, Landt M. Adipose tissue leptin production and plasma leptin kinetics in humans. Diabetes. 1996;45(7):984-987.
    1. Burnett LC, Skowronski AA, Rausch R, LeDuc CA, Leibel RL. Determination of the half-life of circulating leptin in the mouse. Int J Obes (Lond). 2017;41(3):355-359.
    1. Wu X, Motoshima H, Mahadev K, Stalker TJ, Scalia R, Goldstein BJ. Involvement of AMP-activated protein kinase in glucose uptake stimulated by the globular domain of adiponectin in primary rat adipocytes. Diabetes. 2003;52(6):1355-1363.
    1. Fu Y, Luo N, Klein RL, Garvey WT. Adiponectin promotes adipocyte differentiation, insulin sensitivity, and lipid accumulation. J Lipid Res. 2005;46(7):1369-1379.
    1. Morley TS, Xia JY, Scherer PE. Selective enhancement of insulin sensitivity in the mature adipocyte is sufficient for systemic metabolic improvements. Nat Commun. 2015;6:7906.
    1. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89(6):2548-2556.
    1. Manolopoulos KN, Karpe F, Frayn KN. Depot-specific and sex-specific secretion of leptin and interleukin 6: higher leptin release in women and lower interleukin-6 release from femoral adipose tissue in vivo. Lancet. 2013;381:S71.
    1. Trim W, Walhin J, Koumanov F, et al. . Data from “The impact of long-term physical inactivity on adipose tissue immunometabolism”. Bath: University of Bath Research Data Archive. 2021. 10.15125/BATH-01052

Source: PubMed

3
S'abonner