Skip to main content

MINI REVIEW article

Front. Immunol., 21 July 2021
Sec. Nutritional Immunology
This article is part of the Research Topic Aging and Immune Function View all 3 articles

Targeting Aging: Lessons Learned From Immunometabolism and Cellular Senescence

  • Center on Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States

It is well known that aging is associated with dysregulated metabolism. This is seen both in terms of systemic metabolism, as well as at the cellular level with clear mitochondrial dysfunction. More recently, the importance of cellular metabolism in immune cells, or immunometabolism, has been highlighted as a major modifier of immune cell function. Indeed, T cell activation, differentiation, and effector function partly depend on alterations in metabolic pathways with different cell types and functionality favoring either glycolysis or oxidative phosphorylation. While immune system dysfunction with aging is well described, what remains less elucidated is how the integral networks that control immune cell metabolism are specifically affected by age. In recent years, this significant gap has been identified and work has begun to investigate the various ways immunometabolism could be impacted by both chronological age and age-associated symptoms, such as the systemic accumulation of senescent cells. Here, in this mini-review, we will examine immunometabolism with a focus on T cells, aging, and interventions, such as mTOR modulators and senolytics. This review also covers a timely perspective on how immunometabolism may be an ideal target for immunomodulation with aging.

Introduction

As humans age, progressive deterioration leads to impaired function and continual decline that results in increased risk of disease and death (1). Aging is characterized by six hallmarks that contribute to this process and determine the aging phenotype. These include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, altered intracellular communication, stem cell exhaustion, cellular senescence, mitochondrial dysfunction, and dysregulated nutrient sensing (1). Two hallmarks, mitochondrial dysfunction and dysregulated nutrient sensing, are tightly associated with metabolic alterations. Increasing research is investigating the role of metabolism in controlling longevity (2). In line with the hallmarks of aging, several metabolic changes accumulate with age, indicating the presence of a “metabolic clock” (2).

Three metabolic and nutrient sensing pathways, mechanistic target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and sirtuins, are under investigation as potential targets for aging (1, 3). mTOR is involved in key cellular processes such as sensing amino acid concentrations, protein synthesis, and autophagy. mTOR is essential for maintaining physiological and cellular homeostasis, and its dysregulation is associated with various metabolic disorders, cancer, and aging (4). mTOR kinase is involved with two complexes, mTORC1 and mTORC2, that modulate most aspects of anabolic metabolism (5). mTOR activity is associated with aging and longevity and is an attractive investigational target in the aging field (6, 7). AMPK detects low energy states by sensing high AMP levels, and sirtuins detect low energy states by sensing high NAD+ levels (3). These pathways are involved in detecting nutrient scarcity and catabolism, and have age-associated dysregulation (3). Upregulation of AMPK and sirtuins has been shown to favor healthy aging (3). Additionally, AMPK activation has a multitude of effects on metabolism and healthspan and downregulates mTORC1 (8). Various pathways such as the ones above have all been implicated in dysregulated metabolism with aging and may be useful therapeutic targets for age-related deficits.

Although all systems are affected, the hallmarks of aging substantially impact the immune system. It is well known that aging leads to progressive declines in innate and adaptive immunity (912). This immunosenescence is accompanied by chronic low-grade inflammation, or inflammaging (13, 14). This results in increased susceptibility to infections, reduced response to vaccination, and increased prevalence of cancers, autoimmune and chronic diseases. These deficits are reviewed in detail elsewhere (912). Not surprisingly, given that deregulated nutrient sensing and mitochondrial dysfunction are hallmarks of aging (1), markers of inflammaging also coincide with markers of metabolic dysfunction (13). Recent research has highlighted the importance of mitochondrial function and cellular metabolism in controlling immune cell function. Indeed, immunometabolism is critical for proper immune function (1517). What remains to be fully explored is how age and age-associated factors, such as senescent cell accumulation, impact immunometabolism and therefore immune function. This research gap represents a potentially fruitful target for immune modulation in older adults. Here in this mini-review, we will provide an overview of aging, senescence, and immunometabolism with a focus on T cells and interventions, such as mTOR modulators and senolytics.

Immunometabolism

Metabolic control of the immune system provides insight into cellular mechanisms influencing immune cell fate and function. Six major metabolic pathways including glycolysis, fatty acid oxidation (FAO), fatty acid synthesis, tricarboxylic acid (TCA) cycle, amino acid metabolism, and the pentose phosphate pathway are involved in controlling immune cell metabolism (1517). Cell intrinsic and extrinsic signals modulate the activity of immune cells by altering metabolism. Metabolic signaling helps regulate growth, proliferation, and survival needs of immune cells while also influencing effector function. Immune cell subtypes adopt distinct metabolic phenotypes to balance energy requirements, biosynthesis demands, and longevity. During an immune response, immune cells must become activated and change their functional activities in order to respond appropriately (16). Additionally, immune cells must traverse multiple tissues and microenvironments with varying oxygen levels and nutrient availability (18). Thus, immune cells face different metabolic requirements based on their state and tissue environment.

Generally, resting immune cells are relatively inert, but undergo distinct metabolic reprogramming in response to pathogens. Pro-inflammatory immune cells are engaged in aerobic glycolysis where glucose is metabolized into lactate in the presence of oxygen. This glycolytic phenotype is necessary for actively proliferating immune cells to provide biosynthetic precursors essential for synthesis of amino acids, nucleotides, and lipids (19). Additionally, aerobic glycolysis provides carbon intermediates that feed into other biosynthetic pathways and supplies components necessary to sustain proliferation and production of inflammatory molecules (20). Cells with a glycolytic signature can also adapt to restrictive conditions like hypoxia (20). Conversely, immune cells can also utilize the TCA cycle and oxidative phosphorylation (OXPHOS). This pathway is generally employed by immune cells that are quiescent or non-proliferative whose main requirements are energy and longevity (18). In actuality, immune cell subsets use a combination of OXPHOS, glycolysis, and aerobic glycolysis to regulate immune function. The metabolism of many specific immune cell subsets has been previously reviewed (16, 18) and is beyond the scope of this mini-review. In short, metabolism plays multiple roles in various immune cells to regulate immune cell function.

T Cell Immunometabolism

T cells are particularly influenced by metabolic signaling. T cells develop and mature in the thymus and then enter circulation as naïve T cells (Tnaive). Once stimulated with antigen, Tnaive differentiate into effector T (Teff) cells and memory T (Tm) cells. The transition from Tnaive to Teff to Tmem cells requires distinct metabolic coordination. The metabolic events surrounding the coordination of T cell quiescence and activation have been reviewed previously (21, 22). Initially, Tnaive are mostly metabolically inert and generate energy through OXPHOS which is suited for their resting lifestyle. Specifically, Tnaive primarily rely on glucose-derived pyruvate from OXPHOS, or utilize FAO (2326). During T cell activation, there is metabolic reprogramming of antigen specific T cells which have been stimulated via T cell receptor (TCR) ligation and binding of costimulatory molecules. The switch from Tnaive to an activated Teff is characterized by an overall shift in metabolic programing favoring glycolysis over OXPHOS (20). Despite being less efficient than OXPHOS at generating ATP, aerobic glycolysis allows for generation of metabolic intermediates necessary for proliferation and assists in maintaining redox balance (20).

Additionally, CD4 T helper (Th) cell subset differentiation and function is highly dependent on metabolism. Alterations in glycolytic metabolism and mTOR signaling promote differentiation of Th cells into Th1, Th2, Th17, or T regulatory (Treg) cells (2729). Tregs depend more FAO, whereas Th1, Th2, and Th17 cells strongly activate glycolysis through mTOR signaling (29). Changes to Teff cell metabolism, in both the CD4 and CD8 compartments, are critical for the transition of Teff to Tmem, which are generally quiescent and rely on OXPHOS. Subsequently, following antigen challenge, Tmem increase both OXPHOS and glycolysis to facilitate recall responses (3032).

Aging and T Cell Immunometabolism

Metabolic alterations are closely linked to the changes seen in various immune cell populations, including T cells, with aging. This can result in impaired function as well as compromised effectiveness when facing infections (33). The effects of aging on T cell immunity are broad and include both T cell intrinsic and extrinsic effects. With aging, T cell populations shift from mainly naïve to memory subsets (34). Additionally, aging is associated with reduced TCR diversity (35). These phenotypic changes result in overall compromised response to new antigen.

It’s likely that changes to T cell metabolism partially drive these phenotypic differences seen with aging since metabolism is a critical component of effective immune responses. Current research supports metabolic differences between young and aged T cells that can alter cellular function. Specifically, CD4 T cells from older adults had higher baseline and maximal oxygen consumption rate, extracellular acidification rate ratio, and proton leak compared with young CD4 T cells (36). Additionally, upon activation, CD4 T cells from aged mice had altered metabolic reprogramming resulting in compromised metabolic pathways such as OXPHOS and glycolysis (37). Other studies demonstrated metabolic differences such as altered spare respiratory capacity between young and aged CD8 T cells (38). The mechanisms driving metabolic differences in aging T cells are not fully understood, but current research is investigating metabolic profiles in aged T cell subsets and how this impacts function (39). While the research is limited on direct alterations in immunometabolism in aged T cells, many lines of thought support the idea that metabolic changes with aging affect immunometabolism. Future research will better define these changes and how they can be targeted for immune modulation.

Cellular Senescence and Immunometabolism

While work focused on immunometabolism has revealed potentially intervenable deficits associated with age, indirect mechanisms that can also contribute to immune dysfunction. Namely, senescent cells have shown to be key players in the exacerbation of various symptoms of aging and likely contribute to declines in immune cell function and optimal immunometabolism. In the past decade, many studies have linked cellular senescence and senescent cell accumulation with age-related phenotypes and diseases. Cellular senescence is a mostly irreversible state of cell cycle arrest that was initially understood as terminal replicative exhaustion (40). Years later, it became apparent that senescent cells contribute to the overall progression of age-related dysfunction at the organismal level. It was also found that various stimuli induce senescence other than replicative exhaustion. Oncogene expression, genotoxic stress, growth factor signaling, and loss of proteostasis can also induce senescence (41). With age, senescent cells accumulate in various tissues and contribute to tissue dysfunction. Accumulation of these cells also causes a systemic switch in metabolism (42). Although proliferation is arrested, senescent cells are very metabolically active. In general, senescent cells are more glycolytic than their non-senescent counterparts (43). Senescent cells require synthesis of a myriad of signaling molecules related to their secretory program, the senescence associate secretory phenotype (SASP). By increasing reliance on glycolysis, senescent cells are able to support production of this complex and destructive SASP.

The SASP is a heterogeneous, generally pro-inflammatory mix of cytokines, chemokines, and other molecules that contribute to the dysfunction caused by senescent cells (44). These factors alter the tissue microenvironments and induce the chronic, basal, and sterile inflammation, i.e., inflammaging, that is seen with age. Senescent cells, through the SASP, can signal in a paracrine matter to induce senescence in adjacent cells (45). This accumulation of senescent cells will then not only increase the deleterious effects of the SASP but also cause a significant shift in metabolic programming within the tissues (46). Because senescent cells rely heavily on glycolytic pathways, this results in dramatic changes to metabolite utilization and can cause redox stress, further inducing senescence within local environments (47). Although little has been done to assess how this affects the function of immune cells, it is likely that the accumulation of senescent cells and this shift in metabolism negatively impacts immune responses. Indeed, the effect of many different SASP components have been shown to have individual effects on immune cell fate and function (48).

As noted, proper T cell function requires complex orchestration of metabolic switching. While it is controversial if T cells themselves become truly senescent since they tend to lose the ability to secrete cytokines (4852), it is clear that with age, T cells have lower proliferative capacity and altered functionality (4850, 53, 54). Many of the mechanisms that are involved with coordinating their functionality are altered with exposure to SASP factors (46). mTOR is highly active in senescent cells for SASP production (55, 56). Aberrant mTOR activation therefore can be described as an underlying cause and symptom of systemic dysfunction wrought by the accumulation of senescent cells and SASP.

Nicotinamide adenine dinucleotide (NAD+) is another key metabolite that has dramatic changes with age. NAD+ participates in many processes of metabolic homeostasis, including redox reaction steps in nearly every major metabolic pathway. Glycolysis, TCA cycle, OXPHOS, and FAO all include NAD+. With age, however, systemic levels of NAD+ decline and have been linked to many of the hallmarks of aging. Aging causes increased activity of NAD+ consuming pathways including poly (ADP-ribose) polymerases (PARP) mediated DNA-damage repair (57) and sirtuin activity (58), which drive this systemic decrease. CD38, a cell-surface glycoprotein that consumes NAD+ to generate ADP ribose, has emerged as the key regulator of NAD+ decline via a sirtuin-dependent pathway (59). CD38 is highly expressed in a variety of immune cells and participation of immune cells in this paradigm is emerging. Recently, it was shown that culturing macrophages with SASP factors upregulated CD38 expression, which caused a marked decline in systemic NAD+ levels (60). Complicating this paradigm, NAD+ has also been shown to reinforce the secretome of senescent cells via AMPK signaling (61). Aside from the direct role played by macrophages in mediating the decline of NAD+, the effects of this paucity on adaptive immune function have yet to be fully explored. Teff rely heavily on NAD+ to support glutaminolysis and glycolysis, thus NAD+ scarcity could play a large role in T cell functional deficits with age.

Discussion

Interventions that target dysregulated metabolism or senescence may prove fruitful for improving aged immune responses, leading to additional protection when dealing with infection. In fact, Mannick et al. (62) demonstrated that low dose TORC1 inhibition in older adults decreased risk of all infections, upregulated antiviral immunity, and improved influenza vaccination responses. This work confirmed that metabolism is a valuable target to improve immune responses in older adults. Current research is investigating how metabolic regulators affect T cell function and metabolism with aging. Indeed, metformin, an FDA approved diabetes drug that modulates mTOR/AMPK has been investigated as a potential therapeutic to target deficits with T cell aging. Metformin enhances T cell autophagy, normalizes mitochondrial function, and alleviates aging-associated Th17 inflammation (36). Similar to rapamycin, metformin extends healthspan and lifespan in multiple animal models (6366). Additionally, in young mice, metformin can increase CD8 T cell memory formation through AMPK activation and FAO enhancement (31). Although further research is necessary to fully elucidate the mechanisms by which metabolic changes with aging contribute to T cell dysfunction, current metabolic therapeutics may prove beneficial for targeting the aging immune system.

Furthermore, targeting senescent cells may also improve immunometabolism with aging. Senolytics, drugs that target senescent cells, have shown great promise with treating age-related diseases and phenotypes. Treatment with the combination of dasatinib and quercetin (D+Q) is the first described and most well characterized senolytic (67) and improves both life- and healthspan in mice (68). Also in mice, D+Q has treated idiopathic pulmonary fibrosis (69), non-alcoholic fatty liver disease (70), and type 2 diabetes (T2D) (42). In humans, clinical trials have shown efficacy in clearing senescent cells while alleviating symptoms of diabetic kidney disease (71) and idiopathic pulmonary fibrosis (72). Trials are underway utilizing D+Q in other contexts as well as investigating the use of other senolytics, including fisetin. Studies directly linking senolytics and metabolism have been mostly limited to alleviation of T2D. With age, there is increased insulin resistance driven by dysfunction in adipose tissue (73). Senescent adipocyte progenitor cells were found to be the root cause of dysfunction and when cleared with D+Q insulin resistance is reversed (42). As with most senescence associated phenotypes, SASP is the means by which insulin resistance is likely conferred (74). Interestingly, many immune cells (including T cells, B cells, and NKT cells), are thought to exacerbate insulin resistance (75). Taken together, the ablation of SASP using senolytics could possibly reverse the deleterious activity of these immune cells which could be beneficial beyond insulin resistance. Furthermore, senolytics may reduce CD38 expressing macrophages and preserve NAD+ levels with aging, offering a promising strategy to enhance metabolic fitness with age.

Similarly, targeting the SASP directly or indirectly via mitigation of SASP effects may be a valuable way to improve immunometabolism and immune function. Augmentation of NAD+ via supplementation or other indirect methods may improve aging metabolism. Indeed, supplementation studies to increase NAD+ to improve different aspects of metabolism and healthspan show tremendous promise (7678), however the effect on immunometabolism and immune function requires further study. Similarly, CD38 inhibitors via anti-CD38 antibodies, NAD analogs, flavonoids, or others, have shown promise in tumor immunity, aging, and metabolic diseases (7981). More specifically, thiazoloquin(az)olin(on)e CD38 inhibitor can reverse age-related NAD+ decline and improve both functional and metabolic aspects of aging (79). However, its effect on immune function requires further investigation.

Certainly, given the impact of senescent cells on metabolic function and the vast amount of metabolic dysfunction with aging, it is possible that senolytics can modulate immunometabolism and ameliorate age-related immune decline in the context of infection or even vaccination. This strategy is being currently investigated to improve COVID-19 outcomes in an ongoing clinical trial investigating the use of fisetin to mitigate COVID-19 induced cytokine storm in hospitalized patients (NCT04476953). Another group is targeting metabolism via metformin to improve illness in non-hospitalized COVID-19 patients (NCT04510194). Thus, ongoing research will better define the utility of senolytics and metabolic modulators to improve immunometabolism and immune function in the face of infection.

Non-pharmaceutical alternatives, mainly physical exercise and caloric restriction, have been shown to target many of the hallmarks of aging (8284). Exercise prevents diet-induced cellular senescence (85, 86) and many facets of immunosenescence (87). Similarly, caloric restriction extends lifespan and healthspan in multiple species (83, 84, 88), improves metabolism (89), reduces T cell immunosenescence (90), and reduces markers of senescence (91, 92). Thus, while pharmaceuticals are still under development, exercise and caloric restriction are well-supported modulators to improve age-related metabolic and immune dysfunction.

Conclusions

An immune cell’s ability to modulate its metabolism is intimately linked to optimal function. However, this can become compromised due directly to chronological age or indirectly by consequences of age such as accumulation of senescent cells. While there remains much to be uncovered regarding the mechanisms that connect these multifaceted processes, metabolism and senescence have been shown to be intervenable to ameliorate general age-related diseases. Considering how integral the pathways regulating metabolism and senescence are to immune cell function, they are emerging as attractive and efficacious targets. This creates a new possible point of convergence between the worlds of aging, metabolomics, cellular senescence, and immunology that could be of great importance when pursuing strategies to promote healthy aging and a resilient immune system throughout life.

Author Contributions

DM and BT wrote the manuscript. LH and JB helped write and critically reviewed the manuscript. JB finalized the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was funded by R21AG060389 (LH) and R21AG060707 (LH).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The Hallmarks of Aging. Cell (2013) 153(6):1194–217. doi: 10.1016/j.cell.2013.05.039

PubMed Abstract | CrossRef Full Text | Google Scholar

2. López-Otín C, Galluzzi L, Freije JMP, Madeo F, Kroemer G. Metabolic Control of Longevity. Cell Press (2016) p:802–21. doi: 10.1016/j.cell.2016.07.031

CrossRef Full Text | Google Scholar

3. Houtkooper RH, Williams RW, Auwerx J. Metabolic Networks of Longevity. Cell (2010) p:9–14. doi: 10.1016/j.cell.2010.06.029

CrossRef Full Text | Google Scholar

4. Liu GY, Sabatini DM. mTOR at the Nexus of Nutrition, Growth, Ageing and Disease. Nat Res (2020) p:183–203. doi: 10.1038/s41580-019-0199-y

CrossRef Full Text | Google Scholar

5. Laplante M, Sabatini DM. mTOR Signaling in Growth Control and Disease. Cell (2012) p:274–93. doi: 10.1016/j.cell.2012.03.017

CrossRef Full Text | Google Scholar

6. Lamming DW, Ye L, Katajisto P, Goncalves MD, Saitoh M, Stevens DM, et al. Rapamycin-Induced Insulin Resistance is Mediated by mTORC2 Loss and Uncoupled From Longevity. Science (2012) 335(6076):1638–43. doi: 10.1126/science.1215135

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Selman C, Tullet JMA, Wieser D, Irvine E, Lingard SJ, Choudhury AI, et al. Ribosomal Protein S6 Kinase 1 Signaling Regulates Mammalian Life Span. Science (2009) 326(5949):140–4. doi: 10.1126/science.1177221

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Alers S, Loffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the Regulation of Autophagy: Cross Talk, Shortcuts, and Feedbacks. Mol Cell Biol (2012) 32(1):2–11. doi: 10.1128/MCB.06159-11

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Nikolich-Zugich J. The Twilight of Immunity: Emerging Concepts in Aging of the Immune System. Nat Immunol (2018) 19(1):10–9. doi: 10.1038/s41590-017-0006-x

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Solana R, Tarazona R, Gayoso I, Lesur O, Dupuis G, Fulop T. Innate Immunosenescence: Effect of Aging on Cells and Receptors of the Innate Immune System in Humans. Semin Immunol (2012) 24(5):331–41. doi: 10.1016/j.smim.2012.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Shaw AC, Goldstein DR, Montgomery RR. Age-Dependent Dysregulation of Innate Immunity. Nat Rev Immunol (2013) 13(12):875–87. doi: 10.1038/nri3547

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Kogut I, Scholz JL, Cancro MP, Cambier JC. B Cell Maintenance and Function in Aging. Semin Immunol (2012) 24(5):342–9. doi: 10.1016/j.smim.2012.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: A New Immune-Metabolic Viewpoint for Age-Related Diseases. Nat Rev Endocrinol (2018) 14(10):576–90. doi: 10.1038/s41574-018-0059-4

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-Aging. An Evolutionary Perspective on Immunosenescence. Ann N Y Acad Sci (2000) 908:244–54. doi: 10.1111/j.1749-6632.2000.tb06651.x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Norata GD, Caligiuri G, Chavakis T, Matarese G, Netea MG, Nicoletti A, et al. The Cellular and Molecular Basis of Translational Immunometabolism. Immunity (2015) 43(3):421–34. doi: 10.1016/j.immuni.2015.08.023

PubMed Abstract | CrossRef Full Text | Google Scholar

16. O’Neill LA, Kishton RJ, Rathmell J. A Guide to Immunometabolism for Immunologists. Nat Rev Immunol (2016) 16(9):553–65. doi: 10.1038/nri.2016.70

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Palsson-McDermott EM, O’Neill LAJ. Targeting Immunometabolism as an Anti-Inflammatory Strategy. Cell Res (2020) 30(4):300–14. doi: 10.1038/s41422-020-0291-z

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Loftus RM, Finlay DK. Immunometabolism: Cellular Metabolism Turns Immune Regulator. Am Soc Biochem Mol Biol Inc (2016) p:1–10. doi: 10.1074/jbc.R115.693903

CrossRef Full Text | Google Scholar

19. Donnelly RP, Finlay DK. Glucose, Glycolysis and Lymphocyte Responses. Mol Immunol (2015) 68(2):513–9. doi: 10.1016/j.molimm.2015.07.034

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Heiden MGV, Cantley LC, Thompson CB. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science (2009) p:1029–33. doi: 10.1126/science.1160809

CrossRef Full Text | Google Scholar

21. Chapman NM, Boothby MR, Chi H. Metabolic Coordination of T Cell Quiescence and Activation. Nat Res (2020) p:55–70. doi: 10.1038/s41577-019-0203-y

CrossRef Full Text | Google Scholar

22. Buck MD, O’Sullivan D, Pearce EL. T Cell Metabolism Drives Immunity. J Exp Med (2015) 212(9):1345–60. doi: 10.1084/jem.20151159

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Fox CJ, Hammerman PS, Thompson CB. Fuel Feeds Function: Energy Metabolism and the T-Cell Response. Nat Rev Immunol (2005) 5(11):844–52. doi: 10.1038/nri1710

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, et al. The Transcription Factor Myc Controls Metabolic Reprogramming Upon T Lymphocyte Activation. Immunity (2011) 35(6):871–82. doi: 10.1016/j.immuni.2011.09.021

PubMed Abstract | CrossRef Full Text | Google Scholar

25. van der Windt GJ, Pearce EL. Metabolic Switching and Fuel Choice During T-Cell Differentiation and Memory Development. Immunol Rev (2012) 249(1):27–42. doi: 10.1111/j.1600-065X.2012.01150.x

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Pearce EL, Pearce EJ. Metabolic Pathways in Immune Cell Activation and Quiescence. Immunity (2013) 38(4):633–43. doi: 10.1016/j.immuni.2013.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Kopf H, de la Rosa GM, Howard OM, Chen X. Rapamycin Inhibits Differentiation of Th17 Cells and Promotes Generation of Foxp3+ T Regulatory Cells. Int Immunopharmacol (2007) 7(13):1819–24. doi: 10.1016/j.intimp.2007.08.027

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, et al. The Kinase mTOR Regulates the Differentiation of Helper T Cells Through the Selective Activation of Signaling by mTORC1 and mTORC2. Nat Immunol (2011) 12(4):295–303. doi: 10.1038/ni.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting Edge: Distinct Glycolytic and Lipid Oxidative Metabolic Programs Are Essential for Effector and Regulatory CD4+ T Cell Subsets. J Immunol (2011) 186(6):3299–303. doi: 10.4049/jimmunol.1003613

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, et al. mTOR Regulates Memory CD8 T-Cell Differentiation. Nature (2009) 460(7251):108–12. doi: 10.1038/nature08155

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, et al. Enhancing CD8 T-Cell Memory by Modulating Fatty Acid Metabolism. Nature (2009) 460(7251):103–7. doi: 10.1038/nature08097

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Rao RR, Li Q, Odunsi K, Shrikant PA. The mTOR Kinase Determines Effector Versus Memory CD8+ T Cell Fate by Regulating the Expression of Transcription Factors T-Bet and Eomesodermin. Immunity (2010) 32(1):67–78. doi: 10.1016/j.immuni.2009.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Nian Y, Minami K, Maenesono R, Iske J, Yang J, Azuma H, et al. Changes of T-Cell Immunity Over a Lifetime. Transplantation (2019) 103(11):2227–33. doi: 10.1097/TP.0000000000002786

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Koch S, Larbi A, Derhovanessian E, Özcelik D, Naumova E, Pawelec G. Multiparameter Flow Cytometric Analysis of CD4 and CD8 T Cell Subsets in Young and Old People. Immun Ageing (2008) 5(1):6. doi: 10.1186/1742-4933-5-6

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Naylor K, Li G, Vallejo AN, Lee W-W, Koetz K, Bryl E, et al. The Influence of Age on T Cell Generation and TCR Diversity. J Immunol (2005) 174(11):7446–52. doi: 10.4049/jimmunol.174.11.7446

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et al. Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab (2020) 32(1):44–55.e6. doi: 10.1016/j.cmet.2020.04.015

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Nian Y, Iske J, Maenosono R, Minami K, Heinbokel T, Quante M, et al. Targeting Age-Specific Changes in CD4(+) T Cell Metabolism Ameliorates Alloimmune Responses and Prolongs Graft Survival. Aging Cell (2021) 20(2):e13299. doi: 10.1111/acel.13299

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Quinn KM, Hussain T, Kraus F, Formosa LE, Lam WK, Dagley MJ, et al. Metabolic Characteristics of CD8(+) T Cell Subsets in Young and Aged Individuals Are Not Predictive of Functionality. Nat Commun (2020) 11(1):2857. doi: 10.1038/s41467-020-16633-7

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Quinn KM, Palchaudhuri R, Palmer CS, La Gruta NL. The Clock Is Ticking: The Impact of Ageing on T Cell Metabolism. Clin Transl Immunol (2019) 8(11):e01091. doi: 10.1002/cti2.1091

CrossRef Full Text | Google Scholar

40. Hayflick L, Moorhead PS. The Serial Cultivation of Human Diploid Cell Strains. Exp Cell Res (1961) 25(3):585–621. doi: 10.1016/0014-4827(61)90192-6

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Muñoz-Espín D, Serrano M. Cellular Senescence: From Physiology to Pathology. Nat Rev Mol Cell Biol (2014) 15(7):482–96. doi: 10.1038/nrm3823

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, et al. Targeting Senescent Cells Alleviates Obesity-Induced Metabolic Dysfunction. Aging Cell (2019) 18(3):e12950. doi: 10.1111/acel.12950

PubMed Abstract | CrossRef Full Text | Google Scholar

43. James EL, Michalek RD, Pitiyage GN, de Castro AM, Vignola KS, Jones J, et al. Senescent Human Fibroblasts Show Increased Glycolysis and Redox Homeostasis With Extracellular Metabolomes That Overlap With Those of Irreparable DNA Damage, Aging, and Disease. J Proteome Res (2015) 14(4):1854–71. doi: 10.1021/pr501221g

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Coppé J-P, Desprez P-Y, Krtolica A, Campisi J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu Rev Pathol: Mech Dis (2010) 5(1):99–118. doi: 10.1146/annurev-pathol-121808-102144

CrossRef Full Text | Google Scholar

45. Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, et al. A Senescent Cell Bystander Effect: Senescence-Induced Senescence. Aging Cell (2012) 11(2):345–9. doi: 10.1111/j.1474-9726.2012.00795.x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Frasca D, Saada YB, Garcia D, Friguet B. Effects of Cellular Senescence on Metabolic Pathways in non-Immune and Immune Cells. Mech Ageing Dev (2021) 194:111428. doi: 10.1016/j.mad.2020.111428

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Kaplon J, Zheng L, Meissl K, Chaneton B, Selivanov VA, Mackay G, et al. A Key Role for Mitochondrial Gatekeeper Pyruvate Dehydrogenase in Oncogene-Induced Senescence. Nature (2013) 498(7452):109–12. doi: 10.1038/nature12154

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Vicente R, Mausset-Bonnefont AL, Jorgensen C, Louis-Plence P, Brondello JM. Cellular Senescence Impact on Immune Cell Fate and Function. Aging Cell (2016) 15(3):400–6. doi: 10.1111/acel.12455

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Akbar AN, Henson SM. Are Senescence and Exhaustion Intertwined or Unrelated Processes That Compromise Immunity? Nat Rev Immunol (2011) 11(4):289–95. doi: 10.1038/nri2959

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Chou JP, Effros RB. T Cell Replicative Senescence in Human Aging. Curr Pharm design (2013) 19(9):1680–98. doi: 10.2174/138161213805219711

CrossRef Full Text | Google Scholar

51. Crespo J, Sun H, Welling TH, Tian Z, Zou W. T Cell Anergy, Exhaustion, Senescence, and Stemness in the Tumor Microenvironment. Curr Opin Immunol (2013) 25(2):214–21. doi: 10.1016/j.coi.2012.12.003

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Wherry EJ. T Cell Exhaustion. Nat Immunol (2011) 12(6):492–9. doi: 10.1038/ni.2035

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Akbar AN. The Convergence of Senescence and Nutrient Sensing During Lymphocyte Ageing. Clin Exp Immunol (2017) 187(1):4–5. doi: 10.1111/cei.12876

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Akbar AN, Henson SM, Lanna A. Senescence of T Lymphocytes: Implications for Enhancing Human Immunity. Trends Immunol (2016) 37(12):866–76. doi: 10.1016/j.it.2016.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Laberge R-M, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, et al. mTOR Regulates the Pro-Tumorigenic Senescence-Associated Secretory Phenotype by Promoting IL1A Translation. Nat Cell Biol (2015) 17(8):1049–61. doi: 10.1038/ncb3195

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, et al. mTOR Regulates MAPKAPK2 Translation to Control the Senescence-Associated Secretory Phenotype. Nat Cell Biol (2015) 17(9):1205–17. doi: 10.1038/ncb3225

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Mouchiroud L, Houtkooper Riekelt H, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD+/Sirtuin Pathway Modulates Longevity Through Activation of Mitochondrial UPR and FOXO Signaling. Cell (2013) 154(2):430–41. doi: 10.1016/j.cell.2013.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Haigis MC, Sinclair DA. Mammalian Sirtuins: Biological Insights and Disease Relevance. Annu Rev Pathol: Mech Dis (2010) 5(1):253–95. doi: 10.1146/annurev.pathol.4.110807.092250

CrossRef Full Text | Google Scholar

59. Camacho-Pereira J, Tarragó MG, Chini CCS, Nin V, Escande C, Warner GM, et al. CD38 Dictates Age-Related NAD Decline and Mitochondrial Dysfunction Through an SIRT3-Dependent Mechanism. Cell Metab (2016) 23(6):1127–39. doi: 10.1016/j.cmet.2016.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, et al. Senescent Cells Promote Tissue NAD+ Decline During Ageing via the Activation of CD38+ Macrophages. Nat Metab (2020) 2(11):1265–83. doi: 10.1038/s42255-020-00305-3

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Nacarelli T, Lau L, Fukumoto T, Zundell J, Fatkhutdinov N, Wu S, et al. NAD+ Metabolism Governs the Proinflammatory Senescence-Associated Secretome. Nat Cell Biol (2019) 21(3):397–407. doi: 10.1038/s41556-019-0287-4

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Mannick JB, Morris M, Hockey HU, Roma G, Beibel M, Kulmatycki K, et al. TORC1 Inhibition Enhances Immune Function and Reduces Infections in the Elderly. Sci Trans Med (2018) 10(449):1564. doi: 10.1126/scitranslmed.aaq1564

CrossRef Full Text | Google Scholar

63. Anisimov VN, Berstein LM, Egormin PA, Piskunova TS, Popovich IG, Zabezhinski MA, et al. Metformin Slows Down Aging and Extends Life Span of Female SHR Mice. Cell Cycle (2008) 7(17):2769–73. doi: 10.4161/cc.7.17.6625

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cocheme HM, Noori T, et al. Metformin Retards Aging in C. Elegans by Altering Microbial Folate and Methionine Metabolism. Cell (2013) 153(1):228–39. doi: 10.1016/j.cell.2013.02.035

PubMed Abstract | CrossRef Full Text | Google Scholar

65. De Haes W, Frooninckx L, Van Assche R, Smolders A, Depuydt G, Billen J, et al. Metformin Promotes Lifespan Through Mitohormesis via the Peroxiredoxin PRDX-2. Proc Natl Acad Sci U S A (2014) 111(24):E2501–9. doi: 10.1073/pnas.1321776111

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, et al. Metformin Improves Healthspan and Lifespan in Mice. Nat Commun (2013) 4:2192. doi: 10.1038/ncomms3192

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles’ Heel of Senescent Cells: From Transcriptome to Senolytic Drugs. Aging Cell (2015) 14(4):644–58. doi: 10.1111/acel.12344

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics Improve Physical Function and Increase Lifespan in Old Age. Nat Med (2018) 24(8):1246–56. doi: 10.1038/s41591-018-0092-9

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, et al. Cellular Senescence Mediates Fibrotic Pulmonary Disease. Nat Commun (2017) 8(1):14532. doi: 10.1038/ncomms14532

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, et al. Cellular Senescence Drives Age-Dependent Hepatic Steatosis. Nat Commun (2017) 8(1):15691. doi: 10.1038/ncomms15691

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics Decrease Senescent Cells in Humans: Preliminary Report From a Clinical Trial of Dasatinib Plus Quercetin in Individuals With Diabetic Kidney Disease. EBioMedicine (2019) 47:446–56. doi: 10.1016/j.ebiom.2019.08.069

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in Idiopathic Pulmonary Fibrosis: Results From a First-in-Human, Open-Label, Pilot Study. EBioMedicine (2019) 40:554–63. doi: 10.1016/j.ebiom.2018.12.052

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Kahn BB, Flier JS. Obesity and Insulin Resistance. J Clin Invest (2000) 106(4):473–81. doi: 10.1172/JCI10842

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, et al. Targeting Senescent Cells Enhances Adipogenesis and Metabolic Function in Old Age. eLife (2015) 4:e12997. doi: 10.7554/eLife.12997

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Shu CJ, Benoist C, Mathis D. The Immune System’s Involvement in Obesity-Driven Type 2 Diabetes. Semin Immunol (2012) 24(6):436–42. doi: 10.1016/j.smim.2012.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Martens CR, Denman BA, Mazzo MR, Armstrong ML, Reisdorph N, McQueen MB, et al. Chronic Nicotinamide Riboside Supplementation Is Well-Tolerated and Elevates NAD(+) in Healthy Middle-Aged and Older Adults. Nat Commun (2018) 9(1):1286. doi: 10.1038/s41467-018-03421-7

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Canto C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, et al. The NAD(+) Precursor Nicotinamide Riboside Enhances Oxidative Metabolism and Protects Against High-Fat Diet-Induced Obesity. Cell Metab (2012) 15(6):838–47. doi: 10.1016/j.cmet.2012.04.022

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Igarashi M, Miura M, Williams E, Jaksch F, Kadowaki T, Yamauchi T, et al. NAD(+) Supplementation Rejuvenates Aged Gut Adult Stem Cells. Aging Cell (2019) 18(3):e12935. doi: 10.1111/acel.12935

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Tarrago MG, Chini CCS, Kanamori KS, Warner GM, Caride A, de Oliveira GC, et al. A Potent and Specific CD38 Inhibitor Ameliorates Age-Related Metabolic Dysfunction by Reversing Tissue NAD(+) Decline. Cell Metab (2018) 27(5):1081–95.e10. doi: 10.1016/j.cmet.2018.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Hogan KA, Chini CCS, Chini EN. The Multi-Faceted Ecto-Enzyme CD38: Roles in Immunomodulation, Cancer, Aging, and Metabolic Diseases. Front Immunol (2019) 10:1187. doi: 10.3389/fimmu.2019.01187

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Chini C, Hogan KA, Warner GM, Tarrago MG, Peclat TR, Tchkonia T, et al. The Nadase CD38 Is Induced by Factors Secreted From Senescent Cells Providing a Potential Link Between Senescence and Age-Related Cellular NAD(+) Decline. Biochem Biophys Res Commun (2019) 513(2):486–93. doi: 10.1016/j.bbrc.2019.03.199

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Rebelo-Marques A, De Sousa Lages A, Andrade R, Ribeiro CF, Mota-Pinto A, Carrilho F, et al. Aging Hallmarks: The Benefits of Physical Exercise. Front Endocrinol (Lausanne) (2018) 9:258. doi: 10.3389/fendo.2018.00258

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Fontana L, Partridge L, Longo VD. Extending Healthy Life Span–From Yeast to Humans. Science (2010) 328(5976):321–6. doi: 10.1126/science.1172539

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. Caloric Restriction Mimetics Against Age-Associated Disease: Targets, Mechanisms, and Therapeutic Potential. Cell Metab (2019) 29(3):592–610. doi: 10.1016/j.cmet.2019.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Schafer MJ, White TA, Evans G, Tonne JM, Verzosa GC, Stout MB, et al. Exercise Prevents Diet-Induced Cellular Senescence in Adipose Tissue. Diabetes (2016) 65(6):1606–15. doi: 10.2337/db15-0291

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Chen XK, Yi ZN, Wong GT, Hasan KMM, Kwan JS, Ma AC, et al. Is Exercise a Senolytic Medicine? A systematic review. Aging Cell (2021) 20(1):e13294. doi: 10.1111/acel.13294

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Duggal NA, Pollock RD, Lazarus NR, Harridge S, Lord JM. Major Features of Immunesenescence, Including Reduced Thymic Output, Are Ameliorated by High Levels of Physical Activity in Adulthood. Aging Cell (2018) 17(2):e12750. doi: 10.1111/acel.12750

CrossRef Full Text | Google Scholar

88. Mattison JA, Colman RJ, Beasley TM, Allison DB, Kemnitz JW, Roth GS, et al. Caloric Restriction Improves Health and Survival of Rhesus Monkeys. Nat Commun (2017) 8:14063. doi: 10.1038/ncomms14063

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Bruss MD, Khambatta CF, Ruby MA, Aggarwal I, Hellerstein MK. Calorie Restriction Increases Fatty Acid Synthesis and Whole Body Fat Oxidation Rates. Am J Physiol Endocrinol Metab (2010) 298(1):E108–16. doi: 10.1152/ajpendo.00524.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Messaoudi I, Warner J, Fischer M, Park B, Hill B, Mattison J, et al. Delay of T Cell Senescence by Caloric Restriction in Aged Long-Lived Nonhuman Primates. Proc Natl Acad Sci U S A (2006) 103(51):19448–53. doi: 10.1073/pnas.0606661103

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Fontana L, Mitchell SE, Wang B, Tosti V, van Vliet T, Veronese N, et al. The Effects of Graded Caloric Restriction: XII. Comparison of Mouse to Human Impact on Cellular Senescence in the Colon. Aging Cell (2018) 17(3):e12746. doi: 10.1111/acel.12746

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Fontana L, Nehme J, Demaria M. Caloric Restriction and Cellular Senescence. Mech Ageing Dev (2018) 176:19–23. doi: 10.1016/j.mad.2018.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immunometabolism, geroscience, aging, T cell, senescence

Citation: Martin DE, Torrance BL, Haynes L and Bartley JM (2021) Targeting Aging: Lessons Learned From Immunometabolism and Cellular Senescence. Front. Immunol. 12:714742. doi: 10.3389/fimmu.2021.714742

Received: 25 May 2021; Accepted: 08 July 2021;
Published: 21 July 2021.

Edited by:

Richard J. Simpson, University of Arizona, United States

Reviewed by:

Larissa Langhi Prata, Mayo Clinic, United States
Guillaume Spielmann, Louisiana State University, United States

Copyright © 2021 Martin, Torrance, Haynes and Bartley. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jenna M. Bartley, jbartley@uchc.edu

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.