Metabolic consequences of long-term rapamycin exposure on common marmoset monkeys (Callithrix jacchus)

Corinna Ross, Adam Salmon, Randy Strong, Elizabeth Fernandez, Marty Javors, Arlan Richardson, Suzette Tardif, Corinna Ross, Adam Salmon, Randy Strong, Elizabeth Fernandez, Marty Javors, Arlan Richardson, Suzette Tardif

Abstract

Rapamycin has been shown to extend lifespan in rodent models, but the effects on metabolic health and function have been widely debated in both clinical and translational trials. Prior to rapamycin being used as a treatment to extend both lifespan and healthspan in the human population, it is vital to assess the side effects of the treatment on metabolic pathways in animal model systems, including a closely related non-human primate model. In this study, we found that long-term treatment of marmoset monkeys with orally-administered encapsulated rapamycin resulted in no overall effects on body weight and only a small decrease in fat mass over the first few months of treatment. Rapamycin treated subjects showed no overall changes in daily activity counts, blood lipids, or significant changes in glucose metabolism including oral glucose tolerance. Adipose tissue displayed no differences in gene expression of metabolic markers following treatment, while liver tissue exhibited suppressed G6Pase activity with increased PCK and GPI activity. Overall, the marmosets revealed only minor metabolic consequences of chronic treatment with rapamycin and this adds to the growing body of literature that suggests that chronic and/or intermittent rapamycin treatment results in improved health span and metabolic functioning. The marmosets offer an interesting alternative animal model for future intervention testing and translational modeling.

Keywords: animal models; antiaging; healthspan; longevity; nonhuman primate; sirolimus.

Conflict of interest statement

Conflict of interest statement

The authors have no conflict of interests to declare.

Figures

Figure 1. Change in fat mass
Figure 1. Change in fat mass
(A) Change in fat mass at 1 and 2 months, post-dosing, from pre-dosing (month 0) measurement. Squares = control subjects; triangles = rapamycin subjects (mean ± SD); treatment × time interaction, p < 0.0097; difference in month 0 and month 2 mean for rapamycin treated subjects, p < 0.05, Sidak's multiple comparison test. (B) Change in fat mass from pre-dosing measurement for months 1-11 for rapamycin subjects. * treatment effect, F=5.385, p=0.018, Dunnett's multiple comparison test significant, p < 0.05, for month 0 versus months 2, 3, and 5.
Figure 2. Food intake
Figure 2. Food intake
Daily dry matter intake for months 0-12, month 0 is a pre-dosing measurement. Squares = control subjects; triangles = rapamycin subjects (mean ± SD). * treatment effect, F=8.353, p=0.001, Dunnett's multiple comparison test significant, p

Figure 3. Daily activity

Accelerometer counts per…

Figure 3. Daily activity

Accelerometer counts per hour for months 2-13 of dosing. Squares =…

Figure 3. Daily activity
Accelerometer counts per hour for months 2-13 of dosing. Squares = control subjects; triangles= rapamycin subjects (mean ± SD).

Figure 4. Circulating triglyceride

Circulating triglyceride concentrations…

Figure 4. Circulating triglyceride

Circulating triglyceride concentrations for each rapamycin subject for months 0-6, month…

Figure 4. Circulating triglyceride
Circulating triglyceride concentrations for each rapamycin subject for months 0-6, month 0 is a pre-dosing measurement. The solid horizontal line represents the previously established cut-off point for normal triglyceride concentrations in this species.

Figure 5. Metabolic measures

( A )…

Figure 5. Metabolic measures

( A ) Fasting glucose concentration for months 0-8 for rapamycin…

Figure 5. Metabolic measures
(A) Fasting glucose concentration for months 0-8 for rapamycin subjects, month 0 is a pre-dosing measurement. (B) Glucose area under the curve (AUC) for months 0-8 for rapamycin subjects. (C) QuickI index for rapamycin subjects for months 0-8. (D) QuickI index - 1/[log(fasting insulin) + log(fasting glucose)] for months 0 and 2 of dosing, *(F=5.396, p = 0.0453, Sidak's multiple comparison test, p < 0.05 for month 0 control vs rapamycin treated). For all panels squares = control subjects; triangles = rapamycin subjects (mean ± SD).

Figure 6. Immunoblot results

Immunoblot results for…

Figure 6. Immunoblot results

Immunoblot results for the following: adipose triglyceride lipase (ATGL), pyruvate carboxylase…

Figure 6. Immunoblot results
Immunoblot results for the following: adipose triglyceride lipase (ATGL), pyruvate carboxylase (PCB), glucose-6-phosphatase α (G6Pase), glucose-6-phosphate isomerase (GPI), peroxisome proliferator-activated receptor γ (PPARγ), phospho-pyruvate dehydrogenase kinase (p-PDK1), pyruvate dehydrogenase kinase (PDK1), phosphoenol-pyruvate carboxykinase 1 (PCK1), sterol regulatory element-binding protein 1 (SREBP1) corrected by actin. A. Adipose tissue collected at sacrifice following 14 months of rapamycin (black) or control dosing (grey) (mean ± SE) B. Liver tissue collected at sacrifice following 14 months of rapamycin (black) or control dosing (grey) (mean ± SE) *indicates significance p
Similar articles
Cited by
References
    1. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395. - PMC - PubMed
    1. Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, Hejtmancik JF, Nadon N, Strong R, Wood LK, Woodward MA, Miller RA. Rapamycin slows aging in mice. Aging Cell. 2012;11:675–682. - PMC - PubMed
    1. Zhang Y, Bokov A, Gelfond J, Soto V, Ikeno Y, Hubbard G, Diaz V, Sloane L, Maslin K, Treaster S, Réndon S, van Remmen H, Ward W, et al. Rapamycin extends life and health in C57Bl/6 mice. The Journals of Gerontology. Series A, Biological sciences and medical sciences. 2013;69A:119–130. - PMC - PubMed
    1. Fok WC, Chen Y, Bokov A, Zhang Y, Salmon AB, Diaz V, Javors M, Wood WH, 3rd, Zhang Y, Becker KG, Pérez VI, Richardson A. Mice fed rapamycin have an increase in lifespan associated with major changes in the liver transcriptome. PLoS One. 2014;9:e83988. - PMC - PubMed
    1. Halloran J, Hussong SA, Burbank R, Podlutskaya N, Fischer KE, Sloane LB, Austad SN, Strong R, Richardson A, Hart MJ, Galvan V. Chronic inhibition of mammalian target of rapamycin by rapamycin modulates cognitive and non-cognitive components of behavior throughout lifespan in mice. Neuroscience. 2012;223:102–113. - PMC - PubMed
Show all 52 references
Publication types
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3. Daily activity
Figure 3. Daily activity
Accelerometer counts per hour for months 2-13 of dosing. Squares = control subjects; triangles= rapamycin subjects (mean ± SD).
Figure 4. Circulating triglyceride
Figure 4. Circulating triglyceride
Circulating triglyceride concentrations for each rapamycin subject for months 0-6, month 0 is a pre-dosing measurement. The solid horizontal line represents the previously established cut-off point for normal triglyceride concentrations in this species.
Figure 5. Metabolic measures
Figure 5. Metabolic measures
(A) Fasting glucose concentration for months 0-8 for rapamycin subjects, month 0 is a pre-dosing measurement. (B) Glucose area under the curve (AUC) for months 0-8 for rapamycin subjects. (C) QuickI index for rapamycin subjects for months 0-8. (D) QuickI index - 1/[log(fasting insulin) + log(fasting glucose)] for months 0 and 2 of dosing, *(F=5.396, p = 0.0453, Sidak's multiple comparison test, p < 0.05 for month 0 control vs rapamycin treated). For all panels squares = control subjects; triangles = rapamycin subjects (mean ± SD).
Figure 6. Immunoblot results
Figure 6. Immunoblot results
Immunoblot results for the following: adipose triglyceride lipase (ATGL), pyruvate carboxylase (PCB), glucose-6-phosphatase α (G6Pase), glucose-6-phosphate isomerase (GPI), peroxisome proliferator-activated receptor γ (PPARγ), phospho-pyruvate dehydrogenase kinase (p-PDK1), pyruvate dehydrogenase kinase (PDK1), phosphoenol-pyruvate carboxykinase 1 (PCK1), sterol regulatory element-binding protein 1 (SREBP1) corrected by actin. A. Adipose tissue collected at sacrifice following 14 months of rapamycin (black) or control dosing (grey) (mean ± SE) B. Liver tissue collected at sacrifice following 14 months of rapamycin (black) or control dosing (grey) (mean ± SE) *indicates significance p

References

    1. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395.
    1. Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, Hejtmancik JF, Nadon N, Strong R, Wood LK, Woodward MA, Miller RA. Rapamycin slows aging in mice. Aging Cell. 2012;11:675–682.
    1. Zhang Y, Bokov A, Gelfond J, Soto V, Ikeno Y, Hubbard G, Diaz V, Sloane L, Maslin K, Treaster S, Réndon S, van Remmen H, Ward W, et al. Rapamycin extends life and health in C57Bl/6 mice. The Journals of Gerontology. Series A, Biological sciences and medical sciences. 2013;69A:119–130.
    1. Fok WC, Chen Y, Bokov A, Zhang Y, Salmon AB, Diaz V, Javors M, Wood WH, 3rd, Zhang Y, Becker KG, Pérez VI, Richardson A. Mice fed rapamycin have an increase in lifespan associated with major changes in the liver transcriptome. PLoS One. 2014;9:e83988.
    1. Halloran J, Hussong SA, Burbank R, Podlutskaya N, Fischer KE, Sloane LB, Austad SN, Strong R, Richardson A, Hart MJ, Galvan V. Chronic inhibition of mammalian target of rapamycin by rapamycin modulates cognitive and non-cognitive components of behavior throughout lifespan in mice. Neuroscience. 2012;223:102–113.
    1. Stallone G, Infante B, Grandaliano G, Gesualdo L. Management of side effects of sirolimus therapy. Transplantation. 2009;87:S23–S26.
    1. Montalbano M, Neff GW, Yamashiki N, Meyer D, Bettiol M, Slapak-Green G, Ruiz P, Manten E, Safdar K, O'Brien C, Tzakis AG. A retrospective review of liver transplant patients treated with sirolimus from a single center: an analysis of sirolimus-related complications. Transplantation. 2004;78:264–268.
    1. Fang Y, Westbrook R, Hill C, Boparai RK, Arum O, Spong A, Wang F, Javors MA, Chen J, Sun LY, Bartke A. Duration of rapamycin treatment has differential effects on metabolism in mice. Cell Metabolism. 2013;17:456–462.
    1. Fang Y, Bartke A. Prolonged rapamycin treatment led to beneficial metabolic switch. Aging. 2013;5:328–329.
    1. Leontieva OV, Paszkiewicz GM, Blagosklonny MV. Weekly administration of rapamycin improves survival and biomarkers in obese male mice on high-fat diet. Aging Cell. 2014;13:616–622.
    1. Liu Y, Diaz V, Fernandez E, Strong R, Ye L, Baur JA, Lamming DW, Richardson A, Salmon AB. Rapamycin-induced metabolic defects are reversible in both lean and obese mice. Aging (Albany NY) 2014;6:742–754.
    1. Leontieva OV, Pasckiewicz GM, Blagosklonny MV. Comparison of rapamycin schedules in mice on high-fat diet. Cell Cycle. 2014;13:3350–3356.
    1. Leontieva OV, Paszkiewicz G, Demidenko ZN, Blagosklonny MV. Resveratrol potentiates rapamycin to prevent hperinsulinemia and obesity in male mice on high fat diet. Cell Death and Disease. 2013;4:e472.
    1. Grarup N, Andersen G. Gene-environment interactions in the pathogenesis of type 2 diabetes and metabolism. Current Opinion in Clinical Nutrition & Metabolic Care. 2007;10:420–426.
    1. Halford JC, Boyland EJ, Blundell JE, Kirkham TC, Harrold JA. Pharmacological management of appetite expression in obesity. Nature Reviews Endocrinology. 2010;6:255–269.
    1. Jensen T, Kiersgaard M, Sørensen D, Mikkelsen L. Fasting of mice: a review. Laboratory Animals. 2013;47:225–240.
    1. Li S, Zhang HY, Hu CC, Lawrence F, Gallagher KE, Surapaneni A, Estrem ST, Calley JN, Varga G, Dow ER, Chen Y. Assessment of diet-induced obese rats as an obesity model by comparative functional genomics. Obesity. 2008;16:811–818.
    1. Lai M, Chandrasekera PC, Barnard ND. You are what you eat, or are you? The challenges of translating high-fat-fed rodents to human obesity and diabetes. Nutrition & Diabetes. 2014;4:e135.
    1. Ikemoto S, Takahashi M, Tsunoda N, Maruyama K, Itakura H, Ezaki O. High-fat diet-induced hyperglycemia and obesity in mice: differential effects of dietary oils. Metabolism. 1996;45:1539–1546.
    1. Gallou-Kabani C, Vigé A, Gross MS, Rabès JP, Boileau C, Larue-Achagiotis C, Tomé D, Jais JP, Junien C. C57BL/6J and A/J mice fed a high-fat diet delineate components of metabolic syndrome. Obesity. 2007;15:1996–2005.
    1. Warden CH, Fisler JS. Comparisons of diets used in animal models of high-fat feeding. Cell Metabolism. 2008;7:277.
    1. Tardif SD, Mansfield K, Ratnam R, Ross CN, Ziegler TE. The marmoset as a model of aging and age related disease. Institute for Laboratory Animal Research Journal. 2011;52:54–65.
    1. Ross CN, Davis K, Dobek G, Tardif SD. Aging phenotypes of common marmosets (Callithrix jacchus) Journal of Aging Research. 2012;2012:567143.
    1. Tardif SD, Power ML, Ross CN, Rutherford JN, Layne-Colon DG, Paulik MA. Characterization of obese phenotypes in a small nonhuman primate, the common marmoset (Callithrix jacchus) Obesity. 2009;17:1499–1505.
    1. Wachtman LM, Kramer JA, Miller AD, Hachey AM, Curran EH, Mansfield KG. Differential contribution of dietary fat and monosaccharide to metabolic syndrome in the common marmoset (Callithrix jacchus) Obesity. 2011;19:1145–1156.
    1. Power ML, Ross CN, Schulkin J, Tardif SD. The development of obesity begins at an early age in captive common marmosets (Callithrix jacchus) American Journal of Primatology. 2012;74:261–269.
    1. Power ML, Ross CN, Schulkin J, Ziegler TE, Tardif SD. Metabolic consequences of the early onset of obesity in common marmoset monkeys. Obesity. 2013;21:E592–E598.
    1. Ross CN, Power ML, Artavia J, Tardif SD. Relation of food intake behaviors and obesity development in young common marmoset monkeys. Obesity. 2013;21:1891–1899.
    1. Tardif SD, Ross CN, Bergman P, Fernandez E, Javors M, Salmon A, Spross J, Strong R, Richardson A. Testing efficacy of administration of the antiaging drug rapamycin in a nonhuman primate, the common marmoset. Journals of Gerontology: Biological Sciences. 2015;70:577–587.
    1. Verges B, Walter T, Cariou B. Effects of anti-cancer targeted therapies on lipid and glucose metabolism. European Journal of Endocrinology. 2014;170:R43–55.
    1. Mannick JB, Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, Lonetto MA, Maexker HT, Kovarik J, Carson S, Glass DJ, Kickstein LB. mTOR inhibition improves immune function in the elderly. Science Translational Medicine. 2014;268:268ra179.
    1. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006;443:289–295.
    1. Teutonico A, Schena PF, Di Paolo S. Glucose metabolism in renal transplant recipients: effect of calcineurin inhibitor withdrawal and conversion to sirolimus. Journal of the American Society of Nephrology. 2005;16:3128–3135.
    1. Johnston O, Rose CL, Webster AC, Gill JS. Sirolimus is associated with new-onset diabetes in kidney transplant recipients. Journal of the American Society of Nephrology. 2008;197:1411–1418.
    1. Yates CJ, Fourlanos S, Hjelmesaeth J, Colman PG, Cohney SJ. New-onset diabetes after kidney transplantation-changes and challenges. American Journal of Transplantation. 2012;12:820–828.
    1. Pavlakis M, Goldfarb-Rumyantzev AS. Diabetes after transplantation and sirolimus: what's the connection? Journal of the American Society of Nephrology. 2008;19:1255–1256.
    1. Lamming DW, Ye L, Astle CM, Baur JA, Sabatini DM, Harrison DE. Young and old genetically heterogeneous HET3 mice on a rapamycin diet are glucose intolerant but insulin sensitive. Aging Cell. 2013;12:712–718.
    1. Miller RA, Harrison DE, Astle CM, Fernandez E, Flurkey K, Han M, Javors MA, Li X, Nadon NL, Nelson JF, Pletcher S, Salmon AB, Sharp ZD, Van Roekel S, Winkleman L, Strong R. Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell. 2014;13:468–477.
    1. Fraenkel M, Ketzinel-Gilad M, Ariav Y, Pappo O, Karaca M, Castel J, Berthault MF, Magnan C, Cerasi E, Kaiser N, Leibowitz G. mTOR inhibition by rapamycin prevents cell adaptation to hyperglycemia and exacerbates the metabolic state in Type 2 diabetes. Diabetes. 2008;57:945–957.
    1. Blagosklonny MV. Rapamycin-induced glucose intolerance: hunger or starvation diabetes. Cell Cycle. 2011;10:4217–4224.
    1. Blagosklonny MV. Once again on rapamycin-induced insulin resistance and longevity: despite of or owing to. Aging. 2012;4:350–358.
    1. Blagosklonny MV. TOR-centric view on insulin resistance and diabetic complications: perspecitve for endocrinologists and gerontologists. Cell Death and Disase. 2013;4:e964.
    1. Ye L, Widlund AL, Sims CA, Lamming DW, Guan Y, Davis JG, Sabatini DM, Harrison DE, Vang O, Baur JA. Rapamycin doses sufficient to extend lifespan do not compromise muscle mitochondrial content or endurance. Aging. 2013;5:539–550.
    1. Ye L, Varamini B, Lamming DW, Sabatini DM, Baur JA. Rapamycin has a biphasic effect on insulin sensitivity in C2C12 myotubes due to sequential disruption of mTORC1 and mTORC2. Frontiers in Genetics. 2012;3:177.
    1. Lamming DW, Ye L, Katajisto P, Goncalves MD, Saitoh M, Stevens DM, Davis JG, Salmon AB, Richardson A, Ahima RS, Guertin DA, Sabatini DM, Baur JA. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012;335:1638–1643.
    1. Festuccia WT, Blanchard PG, Belchior T, Chimin P, Paschoal VA, Magdalon J, Hirabara SM, Simões D, St-Pierre P, Carpinelli A, Marette A, Deshaies Y. PPARγ activation attenuates glucose intolerance induced by mTOR inhibition with rapamycin in rats. American Journal of Physiology-Endocrinology and Metabolism. 2014;306:E1046–E1054.
    1. Lamming DW, Sabatini DM. A central role for mTOR in lipid homeostasis. Cell Metabolism. 2013;18:465–469.
    1. Sipula IJ, Brown NF, Perdomo G. Rapamycin-mediated inhibition of mammalian target of rapamycin in skeletal muscle cells reduces glucose utilization and increases fatty acid oxidation. Metabolism. 2006;55:1637–1644.
    1. Yu Z, Wang R, Fok WC, Coles A, Salmon AB, Perez VI. Rapamycin and dietary restriction induce metabolically distinctive changes in mouse liver. The Journals of Gerontology. Series A, Biological sciences and medical sciences. 2015;70:410–420.
    1. Houde VP, Brule S, Festuccia WT, Blanchard PG, Bellmann K, Deshaies Y, Marette A. Chronic Rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue. Diabetes. 2010;59:1338–1348.
    1. Layne DG, Power RA. Husbandry, handling, and nutrition for marmosets. Comparative Medicine. 2003;53:351–359.
    1. Ziegler TE, Colman RJ, Tardif SD, Sosa ME, Wegner FH, Wittwer DJ, Shrestha H. Development of metabolic function biomarkers in the common marmoset, Callithrix jacchus. American Journal of Primatology. 2013;75:500–508.

Source: PubMed

3
Subskrybuj