Home » Cores » Aging Animal Models

The Animal Core provides leadership, innovation, facilities and an expertly trained staff to enable investigators to: 1) develop new and study established aging mammalian models; 2) administer drugs and other compounds and provide restricted and other modified diets to elucidate mechanisms and identify treatments for aging; 3) collect longevity and life history data using automated data entry, database acquisition and querying tools. The physical resources, dedicated staff and nationally recognized Leadership ensure the continued ability of the Core to advance discovery of aging mechanisms, with the ultimate goal of translating discovery into therapeutic application to promote and extend the healthy lifespan.

The Animal Core addresses two significant barriers encountered by investigators in aging research by providing: 1) an animal facility dedicated for rodent (mouse and rat) aging studies managed by a staff specifically trained to breed, maintain, administer specific diets and drugs, and conduct lifespan studies of aging rodent colonies and 2) access to animal models of specific interest to biogerontology.

Please be aware that your Protocol must be amended for the service request. Please contact the Core Leaders for more information on protocol modification.

Services Include:

  • Breeding, genotyping, and maintaining mice and rats to any age requested by investigators, which can ensure reproducible, pathogen-free aging animals as well as reduce personnel costs.
  • Conducting lifespan studies of genetically, nutritionally, or pharmacologically manipulated mice and rats.
  • Co-ordination with other cores of the San Antonio Nathan Shock Center (i.e., IPAC, Pathology Core) to provide one-stop-shopping of phenotyping of aging animal.
  • Providing investigators access to aging common marmosets, including tissues and live-animal studies, as a non-human primate model of aging and healthspan.
  • Providing tissue samples and breeding animals from two rodent models of special interest to aging research: naked mole-rats (NMR) and Damaraland mole-rats (DMR).
  • Providing access to “Four Core Genotypes” mice, which are novel tools for exploring the basis for sex differences in lifespan and healthspan.

Recent Key Findings supported by the Animal Core

Identification of the first murine model of the human female survival advantage:
Cheng CJ, Nelson JF. Physiological basis for sex-specific differences in longevity. Current Opinion in Physiology. 2018;6:57-64. doi: https://doi.org/10.1016/j.cophys.2018.04.003.

Discovery that the hyperadreno-corticism caused by calorie restriction mediates resilience against inflammatory stress:
Allen BD, Liao CY, Shu J, Muglia LJ, Majzoub JA, Diaz V, et al. Hyperadrenocorticism of calorie restriction contributes to its anti-inflammatory action in mice. Aging cell. 2019;18(3):e12944. doi: 10.1111/acel.12944. PubMed PMID: 30938024; PubMed Central PMCID: PMC6516174.

Discovery that neural stem cells in the subventricular zone are protected by calorie restriction from age-related deficits:
Apple DM, Mahesula S, Fonseca RS, Zhu C, Kokovay E. Calorie restriction protects neural stem cells from age-related deficits in the subventricular zone. Aging. 2019;11(1):115-26. doi: 10.18632/aging.101731. PubMed PMID: 30622221; PubMed Central PMCID: PMC6339798.

Clarifying changes in autophagy during normal aging in murine muscle:
Zhou J, Chong SY, Lim A, Singh BK, Sinha RA, Salmon AB, et al. Changes in macroautophagy, chaperone-mediated autophagy, and mitochondrial metabolism in murine skeletal and cardiac muscle during aging. Aging. 2017;9(2):583-99. doi: 10.18632/aging.101181. PubMed PMID: 28238968; PubMed Central PMCID: PMC5361683.

Elucidating the role of H2S generation in kidney aging in mice:
Lee HJ, Feliers D, Barnes JL, Oh S, Choudhury GG, Diaz V, et al. Hydrogen sulfide ameliorates aging-associated changes in the kidney. GeroScience. 2018;40(2):163-76. doi: 10.1007/s11357-018-0018-y. PubMed PMID: 29717417; PubMed Central PMCID: PMC5964063.

Characterizing changes with age in kidney, metabolic, cardiovascular and immune function in the marmoset:
Lee HJ, Gonzalez O, Dick EJ, Donati A, Feliers D, Choudhury GG, et al. Marmoset as a Model to Study Kidney Changes Associated With Aging. The journals of gerontology Series A, Biological sciences and medical sciences. 2019;74(3):315-24. doi: 10.1093/gerona/gly237. PubMed PMID: 30321310; PubMed Central PMCID: PMC6376089.

Ross CN, Adams J, Gonzalez O, Dick E, Giavedoni L, Hodara VL, et al. Cross-sectional comparison of health-span phenotypes in young versus geriatric marmosets. American journal of primatology. 2019;81(2):e22952. doi: 10.1002/ajp.22952. PubMed PMID: 30664265.

Establishing the first study of a drug intervention (rapamycin) to extend longevity in marmosets:
Sills AM, Artavia JM, DeRosa BD, Ross CN, Salmon AB. Long-term treatment with the mTOR inhibitor rapamycin has minor effect on clinical laboratory markers in middle-aged marmosets. American journal of primatology. 2019;81(2):e22927. doi: 10.1002/ajp.22927.

Lelegren M, Liu Y, Ross C, Tardif S, Salmon AB. Pharmaceutical inhibition of mTOR in the common marmoset: effect of rapamycin on regulators of proteostasis in a non-human primate. Pathobiology of aging & age related diseases. 2016;6:31793. doi: 10.3402/pba.v6.31793. PubMed PMID: 27341957; PubMed Central PMCID: PMC4920937.

Ross C, Salmon A, Strong R, Fernandez E, Javors M, Richardson A, et al. Metabolic consequences of long-term rapamycin exposure on common marmoset monkeys (Callithrix jacchus). Aging. 2015;7(11):964-73. PubMed PMID: 26568298; PubMed Central PMCID: PMC4694066.

Tardif S, Ross C, Bergman P, Fernandez E, Javors M, Salmon A, et al. Testing efficacy of administration of the antiaging drug rapamycin in a nonhuman primate, the common marmoset. The journals of gerontology Series A, Biological sciences and medical sciences. 2015;70(5):577-87. doi: 10.1093/gerona/glu101. PubMed PMID: 25038772; PubMed Central PMCID: PMC4400395.