World J Mens Health. 2021 Apr;39(2):222-232. English.
Published online Aug 27, 2020.
Copyright © 2021 Korean Society for Sexual Medicine and Andrology
Review

Energy Metabolism and Aging

Andrzej Bartke,1 Savannah Brannan,2 Erin Hascup,3 Kevin Hascup,3 and Justin Darcy4
    • 1Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA.
    • 2Department of Biology, University of Illinois Springfield, Springfield, IL, USA.
    • 3Department of Neurology and Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA.
    • 4Joslin Diabetes Center, Section on Integrative Physiology and Metabolism, Harvard Medical School One Joslin Place, Boston, MA, USA.
Received June 21, 2020; Revised July 04, 2020; Accepted August 28, 2020.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Aging is strongly related to energy metabolism, but the underlying processes and mechanisms are complex and incompletely understood. Restricting energy intake and reducing metabolic rate can slow the rate of aging and extend longevity, implying a reciprocal relationship between energy metabolism and life expectancy. However, increased energy expenditure has also been associated with improved health and longer life. In both experimental animals and humans, reduced body temperature has been related to extended longevity. However, recent findings on the function of thermogenic (brown or beige) adipose tissue produced intense interest in increasing the amount of energy expended for thermogenesis to prevent and/or treat obesity, improve metabolic health, and extend life. Evidence available to-date indicates that increasing adipose tissue thermogenesis by pharmacologic, environmental, or genetic interventions can indeed produce significant metabolic benefits, which are associated with improved chances for healthy aging and long life.

Keywords
Adipose tissue, beige; Adipose tissue, brown; Aging; Metabolic rate; Thermogenesis

INTRODUCTION

All biological functions of living organisms require energy consumption or expenditure. It is, therefore, not surprising that the biological process of aging, and the regulation of longevity, are related in various ways to energy metabolism. Much of the recent and current research on the biology of aging is directed at unraveling these fascinating relationships. This includes studies of the ways in which human energy metabolism could be modified to offset some of the consequences of a socalled “Western” lifestyle in the 21st century, including an imbalance of energy intake and expenditure. In this review article, we will briefly discuss the relationships between metabolic rate and longevity, the impact of various anti-aging interventions on energy metabolism, and the current interest in stimulating thermogenesis to promote healthy aging.

HOW ARE AGING AND LONGEVITY RELATED TO METABOLIC RATE?

In most species of animals, body temperature is identical, or very similar, to environmental temperature (eT). These exothermic (also known as poikilothermic or “cold blooded”) species include worms, snails, mussels, various marine invertebrates, insects, fish, amphibians and reptiles. In these animals, metabolic rate is determined primarily by eT, and thus is low in the cold and high in the warmth. Importantly, in these species, longevity and temperature are inversely related, where exothermic animals live longer in the cold and shorter in the warmth. Naturally, this relationship does not extend to extreme temperatures, which can be detrimental, or even lethal. The inverse relationship between eT and lifespan of exothermic species is striking, consistent, and well documented. This led early investigators to conclude that longevity is determined primarily by metabolic rate. This was long-believed to also apply to endothermic (stenothermic or “warm blooded”) animals, that is, mammals and birds that generally maintain their body temperature within very narrow limits, regardless of eT. This belief was strongly supported by the fact that body mass is a strong determinant of both metabolic rate and longevity with larger species having lower metabolic rate, and longer lifespan, than smaller species [1]. However, there are many striking departures from this relationship [2]. Birds have higher metabolic rates than mammals, but live longer (often much longer) than mammals of the same size. Some mammals (most notably bats and humans) live much longer than their body size would predict. Presumably, the ability to fly (birds and bats), and the intelligence combined with social organization in primates, greatly reduced the risk of death from predation, and thus allowed evolutionary development of life course characteristics and reproductive strategies promoting longer lifespan [3]. Interestingly, both metabolic rate and longevity of humans exceed the corresponding values measured in closely related large apes [4]. This might be related to the greater size and energy requirements of the human brain [4]. Other departures from the simple inverse relationships of metabolic rate and longevity in endotherms include species that deal with extremely low eTs by allowing their body temperature to drop for periods ranging from hours (torpor; common in small rodents) to months (hibernation; i.e., ground squirrels and marmots), and possess multiple physiological adaptations to these states of low metabolic rate [5, 6].

While the concept of mammalian and avian longevity being directly controlled by metabolic rate has been rejected as overly simplistic, some of its elements have been incorporated into the concept of pace-of-life being an important determinant of aging and longevity [7]. Fast pace-of-life includes high metabolic rate, fast postnatal growth, early maturation, high fecundity, and relatively short lifespan, as observed in many species of small rodents. Slow pace-of-life, seen in larger mammals, includes lower metabolic rate, slower growth, later maturation, fewer offspring, and longer life. This concept fits well with the trade-offs between anabolism, growth, and reproduction, on the one hand, and stress resistance, repair processes, and longevity on the other [8, 9]. It also fits well with the role of evolutionarily conserved cellular signaling pathways (insulin, insulin-like growth factors, and mechanistic target of rapamycin) in the control of aging [10] and with the concept of partitioning available energy resources between the processes related to growth and reproduction and those related to maintenance and repair.

WHAT IS THE INFLUENCE OF AGING ON THE METABOLIC RATE?

Progressive decline in physical strength, speed of movements, energy level, and ability to recover from injury, diseases, or environmental insults (resilience) as well as reduced thermogenesis (feeling cold) are among the well-known correlates, and almost certainly effects, of aging. Reduced walking speed and increased difficulty walking for a long distance are among the most consistent functional changes during human aging and are used in geriatric medicine to assess the progression of aging, development of frailty, and risk of neurodegenerative disorders [11, 12, 13]. These observations and age-related decrease in the levels of several anabolic hormones indicate that aging has a major impact on energy metabolism. In fact, an age-related decline in basal metabolic rate was described in both humans [14] and experimental animals [15, 16, 17]. However, there is also evidence that metabolic rate does not always decline with age, and may, in fact, increase due to age-related increases in chronic, low-grade, “sterile” inflammation [18]. This is a result of an accumulation of intra-abdominal adipose tissue that possess an increased abundance of senescent cells, as well as a hypoxic microenvironment, which together, lead to an increased secretion of proinflammatory factors [19]. Thus, age-related changes in metabolic rate in human cohorts representative of the general population can be very different from what is seen in exceptionally healthy individuals [20] in whom inflammation may not override the effects of age on metabolic rate.

ENERGY METABOLISM IS ASSOCIATED WITH EXTREME LONGEVITY

Body temperature is related to metabolic rate, and there is evidence that reduced body temperature favors longevity. Thus, calorie restriction, the most effective way of slowing aging and increasing lifespan of laboratory animals, also results in reduced body temperature in mice [21] as well as in humans [22]. Importantly, this association is very likely to be causal. Bioengineering mice to produce animals with reduced core body temperature increased median lifespan in both females and males [23]. In humans, lower body temperature has been associated with calorie restriction [22] as well as with longevity [24]. A recent publication reported intriguing evidence that mean body temperature in United State adults has been decreasing during the last 157 years by 0.03℃ per birth decade [25]. The authors suggested that this may have contributed to improvements in health and longevity during this period.

We felt that some novel insights into the relationships of aging to energy metabolism can be gained by comparing animals with mutations which predispose them to slow aging and extreme longevity to their genetically normal (wild type) siblings. In mice, various naturally occurring mutations and targeted disruption of genes related to growth hormone (GH) signaling produce syndromes of GH deficiency and GH resistance, which are associated with remarkable extension of longevity in both sexes [reviewed in 8, 26, 27, 28, 29]. We expected to find evidence for reduced metabolism in these long-lived animals, particularly in those mutants which, in addition to being GH-deficient, are also hypothyroid and have a marked reduction in body temperature [30]. However, metabolic rate in these remarkably long-lived mice (assessed by oxygen consumption per unit of body mass) was significantly increased, rather than reduced [31]. We believe that this unexpected finding reflects increased heat loss in these diminutive mutants housed in a standard room temperature of 23℃, and the consequent need for increased thermogenesis. In support of this interpretation, differences of metabolic rate between mutant mice and wild type animals could be diminished or even completely eliminated by housing them in a temperature of 30℃ [32], considered a thermoneutral temperature for mice [33]. In addition to revealing differences in metabolic rate, indirect calorimetry studies of GH-deficient and GH-resistant mice showed a reduction in respiratory quotient (RQ; a ratio of the production of carbon dioxide to consumption of oxygen) [31, 32]. A reduced RQ indicates an increased reliance on fatty acids as opposed to glucose as an energy substrate. Differences in RQ between normal and mutant mice also could be eliminated by housing at increased temperature [32]. An increased usage of lipids as metabolic fuel is associated with improved mitochondrial function, and a reduced production of harmful molecules that cause oxidative stress, both of which are likely mechanisms of aging.

We are currently investigating the effect of long-term exposure to different eTs on aging-sensitive traits and longevity of wild-type and GH-mutant mice. We have recently reported that life-long exposure to increased (thermoneutral) temperature does not eliminate the longevity advantage of GH-resistant Ghr-/- mice [34].

There is increasing evidence that studying mice living at temperatures corresponding to their thermoneutral zone is important for obtaining data that can be “translatable” to humans [35, 36, 37]. Standard animal room temperature is believed to produce conditions of mild cold stress for the mice [33], while most humans live and work in spaces that are heated and/or air-conditioned which, together with wearing weather-appropriate clothing, produces conditions approaching thermoneutrality.

As we mentioned earlier, we hypothesize that in “standard” 23℃ housing temperature (that is cool for mice), the diminutive long-lived GH-related mutants devote a large part of their energy budget to thermogenesis. In support of this hypothesis, Ghr-/-, and Ames dwarf mice have an increased amount of brown adipose tissue (BAT) [38, 39]. Measuring expression of thermogenesis-related genes in this tissue indicates that BAT is more metabolically active in these mutants than in normal animals from the same strain [38, 39]. The role of BAT in thermogenesis, and the potential health benefits of stimulating BAT function, will be discussed in subsequent sections of this article.

BROWN, WHITE, AND BEIGE ADIPOSE TISSUE

Adipose tissue has multiple functions that are critical for health and survival. In contemporary, well-nourished humans, it is the largest organ in the body. In mammals, there are three types of adipose tissue: white adipose tissue (WAT), BAT, and beige adipose tissue [40, 41]. The principal function of WAT is energy storage, which involves accumulation of lipids, which form large lipid droplets in individual white adipocytes. In addition to storing lipids, WAT secretes a multitude of bioactive factors, including leptin, which is important in the regulation of appetite, various proinflammatory molecules, and adiponectin, which enhances sensitivity to insulin and exhibits anti-inflammatory and antiatherogenic activity [42]. WAT is widely distributed, but most of it is present beneath the skin (subcutaneous WAT) and in the abdominal cavity (visceral WAT).

Technological and economic development during the twentieth and twenty-first century led to conditions which promote accumulation of WAT, leading to obesity, which now afflicts more than a quarter of adults in developed nations. Although genetic predisposition plays a role, these conditions essentially reflect a shift in energy metabolism, involving an unequal balance of energy expenditure and consumption. Inflammation of WAT is a result of this imbalance, and it leads to increased risk of type 2 diabetes, cancer, and cardiovascular disease [19, 43, 44]. There is increasing evidence that obesity also increases the risk of Alzheimer's disease and other age-related neuropathology [45, 46, 47, 48, 49], and likely accelerates the rate of aging [50]. These effects of obesity certainly increase mortality rate, and represent one of the key public health issues of the modern world. In this context, the potential of preventing and/or treating obesity by increasing thermogenic functions of adipose tissue is a focus of intense ongoing research.

BAT has a higher concentration of mitochondria than WAT and is responsible for non-shivering thermogenesis. BAT uses thermogenin, also known as uncoupling protein-1 (UCP1), to uncouple the electron transport chain from ATP production, resulting in the production of chemical energy in the form of heat [51]. To maintain this futile process, BAT must consume and expend energy from circulating glucose and lipids. Indeed, while BAT in mice may constitute only a few hundred milligrams, it can consume more than half of ingested lipids and glucose when maximally stimulated [52]. In humans, BAT was once thought to be present only in newborns, and would disappear during adolescence. Studies have now shown that BAT is still present in adult humans [53, 54, 55, 56, 57]. BAT can be found surrounding the circulatory system to dissipate heat, and can be found near the heart, lungs, and kidney. In humans, the largest depot of BAT can be found in the supraclavicular region [56, 58, 59]. Fifty grams of BAT mass accounts for 3% to 5% of a human subject's basal metabolic rate [60]. In mice, BAT differs anatomically from human BAT because it is concentrated in specific depots (e.g., the interscapular depot), whereas in humans, brown adipocytes tend to be dispersed within white adipose depots.

The spatial (anatomical) and functional separation of BAT and WAT is not absolute. Under some circumstances (for example, prolonged cold exposure), white adipocytes can express UCP1, acquire other characteristics of brown adipocytes, and assume a role in thermogenesis and maintenance of body temperature. This process is referred to as “beiging,” and the resulting type of adipose tissue is classified as beige [41, 52]. Beige adipose tissue is sometimes referred to as brite. It is also possible for BAT cells to lose their thermogenic properties and to start resembling white adipocytes. This is called whitening of BAT. Physiologically, BAT and beige fat have widely overlapping (if not identical) functions, and thus, beiging of white fat and stimulation of BAT and beige adipocytes function are expected to produce nearly identical metabolic benefits [61]. It has been suggested that human BAT corresponds more closely to beige rather than brown fat in the mouse, but this is still a matter of some debate [57]. Interestingly, BAT development and metabolic health can be shaped during fetal development by maternal exercise [62].

BENEFITS OF VARIOUS MEANS OF STIMULATING BROWN ADIPOSE TISSUE FUNCTION AND PROMOTING BEIGING OF WHITE ADIPOSE TISSUE

Age- and obesity-related declines in BAT mass and its thermogenic activity are associated with an increase in WAT, and this association is almost certainly causal. In humans, BAT mass has been shown to be inversely correlated with body mass index (BMI) [54], and thus increase in adiposity and reduction of BAT may form a vicious circle [56, 58]. Moreover, BAT levels decline with age [58], although this can be prevented through interventions that stimulate the beiging of fat [63]. Indeed, the decrease in BAT as a result of increased BMI and age raises the possibility that promoting BAT may combat both obesity and age-related metabolic disorders. Methods of improving BAT thermogenesis and promoting formation of beige fat, along with some of the mechanisms which control the function of these tissues, are discussed below. Understanding these mechanisms should identify targets for development of pharmacological interventions.

1. Cold exposure

Cold exposure has been shown to directly increase energy expenditure. The results of a recent study of the effects of outdoor temperatures showed that the energy expenditure and the skin temperature of the supraclavicular region increased with cold exposure [59]. This indicates that BAT concentrated in that region effectively induced non-shivering thermogenesis to produce heat, thereby increasing energy expenditure. It was recently reported that presence of active BAT determines cold-induced increase in energy expenditure in humans [64]. Cold exposure has also been proven to induce white fat beiging [65]. This can reverse the deterioration of BAT with age due to oxidative stress [65], which holds potential in treating metabolic disorders. In mice fed a high fat diet, chronic exposure to low temperature reduced obesity by enhancing BAT thermogenesis [66]. Importantly, intermittent exposure to cold was shown to improve control of blood glucose (“sugar”) levels in patients with type 2 diabetes [67]. In this study, acclimating patients to 15℃, over a 10-day period, while wearing only light clothing resulted in a 43% increase in insulin sensitivity. These, and similar findings, led to suggestions that lower setting of temperature controls in the house, sleeping under lighter covers, having windows open during the night, and using other means of reducing ambient temperature and stimulating thermogenesis are likely to have beneficial effects on body composition and metabolic health [68, 69, 70, 71].

Additional evidence for beneficial effects of cold exposure was obtained from studies of individuals practicing winter (cold water) swimming, an extreme sport which is increasingly popular in various countries [72]. Practitioners of this sport swim regularly in natural bodies of water through most, or all, of winter in locations where water temperature drops to 1℃ to 5℃. Winter swimming produces a host of physiological responses including regional changes in blood flow, acute responses to the stress of cold water immersion (including transient increases in sympathetic tone and cortisol levels), adaptation to this stress, and stimulation of both shivering and non-shivering thermogenesis. Beneficial metabolic effects include improved insulin sensitivity and reduced insulin levels [73, 74], as well as a reduction of and enhanced activity of anti-oxidant enzymes [75]. Although effects of increased thermogenesis in cold water swimmers are difficult (if not impossible) to separate from the effects of exercise and the hormetic responses to repeated stress, the observed physiological changes would be expected to reduce risk of chronic disease and to promote healthy aging. A pronounced reduction in the insulin levels and enhanced insulin sensitivity [73, 74] are of particular note in this context. For the readers who might be interested in starting cold water swimming, we should add that this can be risky, particularly for those with some pre-existing heart conditions and generally untrained individuals without prior adaptation to cold [72, 76]. Please consult with your physician first!

It appears that the potential benefits of immersion in cool water, rather than extremely cold water, have not been extensively explored. Although intensive exercise produces heat, therefore negating the effects of cold exposure, relaxed swimming in cooler water could potentially stimulate non-shivering thermogenesis due to water's evaporative cooling effect.

2. Dietary polyphenols

Polyphenols are micronutrients with multiple phenol groups. Some polyphenols have antioxidant properties, and can benefit metabolic health by targeting the sympathetic nervous system (SNS), which is responsible for adrenergic signaling. The SNS is importantly involved in thermoregulatory processes because it signals to interscapular BAT to induce heat production through activation UCP1 [77]. Various kinds of natural polyphenols can induce SNS signaling, and therefore BAT stimulation. Naringenin is a citrus flavonoid that was tested in human white adipocyte tissue cultures (hADSC) to determine if it induces browning. Studies of Rebello et al [78] showed that naringenin increased the expression of UCP1 and adipose triglyceride lipase, which are both involved in thermogenesis. Oxygen consumption rates increased in naringenin-treated hADSC, signifying that it caused an increase in energy expenditure. Naringenin also increased glucose transporter type 4, adiponectin, and carbohydrate-responsive element-binding protein levels, which are all associated with improving insulin sensitivity [78]. These findings suggest that naringenin could potentially be used for treating obesity and diabetes.

Resveratrol is a polyphenol found in red cabbage, spinach, berries, red grapes and wine, and peanuts. In addition to its impact on sirtuins and on metabolism of carbohydrates and lipids [79, 80], resveratrol can have beneficial effects on the gut microbiome [66]. It can improve gut function and activate metabolites in the intestinal tract through gut microflora mediated-biotransformation [66]. These actions can increase the beiging of WAT tissue.

Green tea extract which contains epigallocatechin gallate can promote weight loss by stimulating thermogenesis. This effect is most likely due to inhibition of the degradation of norepinephrine and the resulting increase in the sympathetic stimulation of BAT [81].

Many other natural plant-derived polyphenols, including thyme and chysin, have been shown to enhance lipolysis [63]. Magnolol and Honokiol are components of bark extracts from Asian trees that have been used for medicinal purposes and have been found to induce thermogenesis, leading to increased lipolysis and reduced lipogenesis [63]. These natural sources of dietary polyphenols may offer possible safe treatment option for inducing lipolysis in obese patients by stimulating the SNS and initiating the beiging of WAT.

Dietary polyphenols combined with cold exposure is another potential treatment method for increasing energy expenditure in obese patients. Capsinoids are known to cause thermogenic activation, increasing energy expenditure through recruiting active BAT during cold exposure [63]. This method has been shown to be effective in patients with low BAT levels as well, meaning this treatment could combat BAT degeneration due to aging. Capsinoid food sources include chili peppers such as jalapeno and cayenne. Coupling a natural dietary polyphenol supplement with cold exposure could offer a safe and natural way to stimulate WAT beiging and raising energy expenditure levels in obese individuals.

3. Oxidative stress

The amount of BAT and its thermogenic functions tend to decline during aging [58]. Oxidative stress is believed to be one of the key mechanisms of aging [82]. The role of oxidative stress in age-related changes in BAT was tested through exposing BAT cells to hydrogen peroxide, an oxidative agent. This caused a decline in BAT cells. Importantly, it was also shown that antioxidant treatment reduced the effects of oxidative stress and reversed BAT activity decline [60]. Mitochondrial dysfunction can develop during aging, causing a decrease in fuel oxidation overall and a buildup of reactive oxidative species [60]. These reactive species are likely responsible for an increase in metabolic dysfunction rates with age, but antioxidant supplements could reduce these effects and reverse BAT deterioration.

4. Endogenous compounds: lipokines and bile acids

Although drugs such as mirabegron are appealing since they can activate BAT, they tend to have undesirable off-target effects, such as tachycardia. Therefore, there has been an emphasis on finding physiological compounds that can target, and activate, BAT. Lipokines, or lipids that are secreted from adipose tissue and have signaling properties, are a novel class of lipid, some of which have been able to increase BAT function. For example, 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) is secreted from activated BAT, and acts in an autocrine manner to stimulate lipid uptake into brown adipocytes [83]. Another example is 12-hydroxyeicosapentaenoic acid (12-HEPE), which is secreted from BAT and acts in an endocrine manner to increase glucose uptake [84]. While it does act as an endocrine lipokine, it is also able to stimulate BAT-specific uptake of glucose as well. Therefore, treatment with 12,13-diHOME and 12-HEPE has the potential to decrease the abundance of circulating lipids and glucose through disposal by BAT. Unfortunately, there is currently no available data on the effects of these lipids on lifespan.

Another type of endogenous compound that can stimulate BAT are bile acids. Bile acids normally function in the gastrointestinal tract to emulsify consumed fat, however, they have garnered considerable attention as endocrine molecules in recent years, particularly in terms of glucose metabolism [reviewed in 85]. Interestingly, several studies have suggested that bile acids are able to stimulate BAT activation, and even protect mice from diet-induced obesity [86, 87]. Interestingly, bile acids have been shown to play a beneficial role in aged animals, particularly in the context of glucose metabolism [88]. Although an attempt to extend longevity through the use of a particular bile acid, ursodeoxycholic acid was unsuccessful [89], there does appear to be a role of bile acids and aging. For example, the long-lived “little” mice have drastically higher levels of many bile acids [90], which appears to be at least partially controlled by growth hormone signaling in the liver [91]. Moreover, treating progeroid mice with cholic acid was able to significantly extend longevity [92]. To what extent bile acids can impact BAT and longevity is still unknown.

STUDIES OF THE REGULATION OF DEVELOPMENT AND FUNCTION OF BROWN AND BEIGE FAT SUGGEST NOVEL TARGETS FOR INTERVENTION

Thermogenesis involves shuttling fatty acids to BAT or beige fat cells to produce heat rather than storing them in WAT [51]. This process in BAT is activated during cold exposure [51, 93]. Targeting this mechanism of fatty acid redistribution could be beneficial for treating obesity and accompanying metabolic diseases, such as cardiovascular disease and diabetes and have been related to excess triglyceride storage in WAT.

Central nervous system control of BAT thermogenesis involves guanylate cyclase 2C receptors in the hypothalamus and it was recently shown that activation of these receptors by treatment with linaclotide reduced body weight of obese mice without changing their food intake [94]. This beneficial metabolic effect was related to inducing increased energy expenditure for BAT thermogenesis. Linaclotide is a drug approved for treatment of irritable bowel syndrome with constipation.

Research in this field is very active and several novel approaches to stimulating energy expenditure in order to improve metabolic health have been reported while this article was being finalized. Treating mice with an orally bioavailable mitochondrial uncoupler, BAM15, was shown to increase nutrient oxidation, and decrease adiposity without altering food intake, and improve insulin sensitivity [95]. These actions reversed diet-induced obesity and would be expected to promote healthy aging. A derivative of amphetamine modified to prevent its entry into the brain and acting as sympathofacilitator was reported to increase lipolysis, thermogenesis, and heat dissipation in mice, thus protecting the animals from obesity without having undesirable cardiovascular and central nervous system effects of amphetamine [96]. Disulfiram, which is approved treatment of alcohol addiction, was recently shown to reverse diet-induced obesity and metabolic dysfunction in mice by increasing energy expenditure [97]. An implantable wireless optogenetic device was shown to activate thermogenesis selectively in adipocytes by a mechanism independent of beta-adrenergic pathway [98]. In mice, this innovative treatment could increase thermogenesis and whole-body energy expenditure and prevent diet-induced weight gain [98]. Results of these and other recent studies identify new avenues that could be explored to stimulate activity of BAT and beige fat in people to produce metabolic changes that reduce a risk of age-related disease likely leading to extended longevity.

CONCLUSIONS

Age-related changes in body composition and in the risk of diabetes and other chronic diseases are related to energy metabolism. Activation of BAT and conversion of WAT to thermogenic beige fat can increase utilization of fat for thermogenesis (heat production) leading to reduced adiposity, improved glucose homeostasis, and healthier metabolism. Increasing energy expenditure for thermogenesis by cold exposure or by dietary or pharmacological interventions holds promise for reducing and/or preventing obesity and improving chances for healthy aging and long life.

Notes

Conflict of Interest:The authors have no potential conflicts of interest to disclose.

Author Contribution:

  • Article conception & design: AB.

  • Data acquisition: AB.

  • Data analysis and interpretation: AB.

  • Writing of the manuscript: AB, KH, EH, SB, JD.

  • Receiving grant: AB, EH, JD.

  • Approval of final manuscript: AB.

ACKNOWLEDGEMENTS

We apologize to those whose work pertinent to the issues discussed was not cited due to limitations of the format or to inadvertent omissions. We are grateful for editorial assistance provided by Lisa Hensley and Dr. Tracy Evans

Writing of this article and our recent and current studies of this topic were supported by American Diabetes Association [ADA 1-19-IBS-126] and the William E. McElroy Charitable Foundation (Bartke A), National Institutes of Health [NIA R01AG057767 and NIA R01AG061937], the Center for Alzheimer's Disease and Related Disorders, and the Kenneth Stark Endowment (Hascup E, Hascup K), and the American Heart Association AHA 20POST35210497 (Darcy J).

References

    1. Pearl R. In: The rate of living. London: University of London Press; 1928.
    1. Austad SN. Cats, “rats,” and bats: the comparative biology of aging in the 21st century. Integr Comp Biol 2010;50:783–792.
    1. Holmes DJ, Austad SN. Birds as animal models for the comparative biology of aging: a prospectus. J Gerontol A Biol Sci Med Sci 1995;50:B59–B66.
    1. Pontzer H, Brown MH, Raichlen DA, Dunsworth H, Hare B, Walker K, et al. Metabolic acceleration and the evolution of human brain size and life history. Nature 2016;533:390–392.
    1. Wu CW, Storey KB. Life in the cold: links between mammalian hibernation and longevity. Biomol Concepts 2016;7:41–52.
    1. Hulbert AJ. Metabolism and longevity: Is there a role for membrane fatty acids? Integr Comp Biol 2010;50:808–817.
    1. Glazier DS. Is metabolic rate a universal 'pacemaker' for biological processes? Biol Rev Camb Philos Soc 2015;90:377–407.
    1. Bartke A, Sun LY, Longo V. Somatotropic signaling: trade-offs between growth, reproductive development, and longevity. Physiol Rev 2013;93:571–598.
    1. Maklakov AA, Chapman T. Evolution of ageing as a tangle of trade-offs: energy versus function. Proc Biol Sci 2019;286:20191604
    1. Bitto A, Wang AM, Bennett CF, Kaeberlein M. Biochemical genetic pathways that modulate aging in multiple species. Cold Spring Harb Perspect Med 2015;5:a025114
    1. Best JR, Liu-Ambrose T, Metti AL, Rosso AL, Satterfield S, Studenski S, et al. Health, Aging and Body Composition Study. Health, Aging and Body Composition Study. Longitudinal associations between walking speed and amount of self-reported time spent walking over a 9-year period in older women and men. J Gerontol A Biol Sci Med Sci 2018;73:1265–1271.
    1. Blaum CS, Xue QL, Tian J, Semba RD, Fried LP, Walston J. Is hyperglycemia associated with frailty status in older women? J Am Geriatr Soc 2009;57:840–847.
    1. Hoogendijk EO, Afilalo J, Ensrud KE, Kowal P, Onder G, Fried LP. Frailty: implications for clinical practice and public health. Lancet 2019;394:1365–1375.
    1. Kitazoe Y, Kishino H, Tanisawa K, Udaka K, Tanaka M. Renormalized basal metabolic rate describes the human aging process and longevity. Aging Cell 2019;18:e12968
    1. Speakman JR, van Acker A, Harper EJ. Age-related changes in the metabolism and body composition of three dog breeds and their relationship to life expectancy. Aging Cell 2003;2:265–275.
    1. Moe B, Rønning B, Verhulst S, Bech C. Metabolic ageing in individual zebra finches. Biol Lett 2009;5:86–89.
    1. Schefer V, Talan MI. Oxygen consumption in adult and AGED C57BL/6J mice during acute treadmill exercise of different intensity. Exp Gerontol 1996;31:387–392.
    1. Fabbri E, An Y, Schrack JA, Gonzalez-Freire M, Zoli M, Simonsick EM, et al. Energy metabolism and the burden of multimorbidity in older adults: results from the Baltimore Longitudinal Study of Aging. J Gerontol A Biol Sci Med Sci 2015;70:1297–1303.
    1. Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, et al. Fat tissue, aging, and cellular senescence. Aging Cell 2010;9:667–684.
    1. Schrack JA, Knuth ND, Simonsick EM, Ferrucci L. “IDEAL” aging is associated with lower resting metabolic rate: the Baltimore Longitudinal Study of Aging. J Am Geriatr Soc 2014;62:667–672.
    1. Green CL, Mitchell SE, Derous D, Wang Y, Chen L, Han JJ, et al. The effects of graded levels of calorie restriction: XIV. global metabolomics screen reveals brown adipose tissue changes in amino acids, catecholamines, and antioxidants after short-term restriction in C57BL/6 mice. J Gerontol A Biol Sci Med Sci 2020;75:218–229.
    1. Conti B, Sanchez-Alavez M, Winsky-Sommerer R, Morale MC, Lucero J, Brownell S, et al. Transgenic mice with a reduced core body temperature have an increased life span. Science 2006;314:825–828.
    1. Heilbronn LK, de Jonge L, Frisard MI, DeLany JP, Larson-Meyer DE, Rood J, et al. ; Pennington CALERIE Team. Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial. JAMA 2006;295:1539–1548.
    1. Roth GS, Lane MA, Ingram DK, Mattison JA, Elahi D, Tobin JD, et al. Biomarkers of caloric restriction may predict longevity in humans. Science 2002;297:811.
    1. Protsiv M, Ley C, Lankester J, Hastie T, Parsonnet J. Decreasing human body temperature in the United States since the industrial revolution. Elife 2020;9:e49555
    1. Bartke A. Growth hormone and aging: updated review. World J Mens Health 2019;37:19–30.
    1. Aguiar-Oliveira MH, Bartke A. Growth hormone deficiency: health and longevity. Endocr Rev 2019;40:575–601.
    1. Brown-Borg HM, Bartke A. GH and IGF1: roles in energy metabolism of long-living GH mutant mice. J Gerontol A Biol Sci Med Sci 2012;67:652–660.
    1. Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ. The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol 2013;9:366–376.
    1. Hunter WS, Croson WB, Bartke A, Gentry MV, Meliska CJ. Low body temperature in long-lived Ames dwarf mice at rest and during stress. Physiol Behav 1999;67:433–437.
    1. Westbrook R, Bonkowski MS, Strader AD, Bartke A. Alterations in oxygen consumption, respiratory quotient, and heat production in long-lived GHRKO and Ames dwarf mice, and short-lived bGH transgenic mice. J Gerontol A Biol Sci Med Sci 2009;64:443–451.
    1. Darcy J, McFadden S, Fang Y, Berryman DE, List EO, Milcik N, et al. Increased environmental temperature normalizes energy metabolism outputs between normal and Ames dwarf mice. Aging (Albany NY) 2018;10:2709–2722.
    1. Gordon CJ. In: Temperature regulation in laboratory rodents. Cambridge: Cambridge University Press; 1993.
    1. Fang Y, McFadden S, Darcy J, Hascup ER, Hascup KN, Bartke A. Lifespan of long-lived growth hormone receptor knockout mice was not normalized by housing at 30°C since weaning. Aging Cell 2020;19:e13123
    1. Fischer AW, Cannon B, Nedergaard J. Optimal housing temperatures for mice to mimic the thermal environment of humans: an experimental study. Mol Metab 2018;7:161–170.
    1. Kajimura S, Spiegelman BM. Confounding issues in the “humanized” BAT of mice. Nat Metab 2020;2:303–304.
    1. de Jong JMA, Sun W, Pires ND, Frontini A, Balaz M, Jespersen NZ, et al. Human brown adipose tissue is phenocopied by classical brown adipose tissue in physiologically humanized mice. Nature Metabolism 2019;1:830–843.
    1. Darcy J, Bartke A. Functionally enhanced brown adipose tissue in Ames dwarf mice. Adipocyte 2017;6:62–67.
    1. Li Y, Knapp JR, Kopchick JJ. Enlargement of interscapular brown adipose tissue in growth hormone antagonist transgenic and in growth hormone receptor gene-disrupted dwarf mice. Exp Biol Med (Maywood) 2003;228:207–215.
    1. Hussain MF, Roesler A, Kazak L. Regulation of adipocyte thermogenesis: mechanisms controlling obesity. FEBS J. 2020 [doi: 10.1111/febs.15331]
      [Epub].
    1. Zwick RK, Guerrero-Juarez CF, Horsley V, Plikus MV. Anatomical, physiological, and functional diversity of adipose tissue. Cell Metab 2018;27:68–83.
    1. Trayhurn P, Beattie JH. Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc 2001;60:329–339.
    1. Barroso TA, Marins LB, Alves R, Souza Gonçalves AC, Barroso SG, de Souza Rocha G. Association of central obesity with the incidence of cardiovascular diseases and risk factors. Int J Cardiovasc Sci 2017;30:416–424.
    1. Ryder JR, Northrop E, Rudser KD, Kelly AS, Gao Z, Khoury PR, et al. Accelerated early vascular aging among adolescents with obesity and/or type 2 diabetes mellitus. J Am Heart Assoc 2020;9:e014891
    1. Valcarcel-Ares MN, Tucsek Z, Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, et al. Obesity in aging exacerbates neuroinflammation, dysregulating synaptic function-related genes and altering eicosanoid synthesis in the mouse hippocampus: potential role in impaired synaptic plasticity and cognitive decline. J Gerontol A Biol Sci Med Sci 2019;74:290–298.
    1. Bischof GN, Park DC. Obesity and aging: consequences for cognition, brain structure, and brain function. Psychosom Med 2015;77:697–709.
    1. Baumgart M, Snyder HM, Carrillo MC, Fazio S, Kim H, Johns H. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: a population-based perspective. Alzheimers Dement 2015;11:718–726.
    1. Palmer AK, Kirkland JL. Aging and adipose tissue: potential interventions for diabetes and regenerative medicine. Exp Gerontol 2016;86:97–105.
    1. Hascup ER, Broderick SO, Russell MK, Fang Y, Bartke A, Boger HA, et al. Diet-induced insulin resistance elevates hippocampal glutamate as well as VGLUT1 and GFAP expression in AβPP/PS1 mice. J Neurochem 2019;148:219–237.
    1. Jura M, Kozak LP. Obesity and related consequences to ageing. Age (Dordr) 2016;38:23
    1. Dijk W, Heine M, Vergnes L, Boon MR, Schaart G, Hesselink MK, et al. ANGPTL4 mediates shuttling of lipid fuel to brown adipose tissue during sustained cold exposure. Elife 2015;4:e08428
    1. Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med 2011;17:200–205.
    1. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med 2009;360:1500–1508.
    1. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med 2009;360:1509–1517.
    1. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional brown adipose tissue in healthy adults. N Engl J Med 2009;360:1518–1525.
    1. Sacks H, Symonds ME. Anatomical locations of human brown adipose tissue: functional relevance and implications in obesity and type 2 diabetes. Diabetes 2013;62:1783–1790.
    1. Cannon B, de Jong JMA, Fischer AW, Nedergaard J, Petrovic N. Human brown adipose tissue: classical brown rather than brite/beige? Exp Physiol. 2020 [doi: 10.1113/EP087875]
      [Epub].
    1. Pfannenberg C, Werner MK, Ripkens S, Stef I, Deckert A, Schmadl M, et al. Impact of age on the relationships of brown adipose tissue with sex and adiposity in humans. Diabetes 2010;59:1789–1793.
    1. Leitner BP, Huang S, Brychta RJ, Duckworth CJ, Baskin AS, McGehee S, et al. Mapping of human brown adipose tissue in lean and obese young men. Proc Natl Acad Sci U S A 2017;114:8649–8654.
    1. Cui X, Xiao W, You L, Zhang F, Cao X, Feng J, et al. Age-induced oxidative stress impairs adipogenesis and thermogenesis in brown fat. FEBS J 2019;286:2753–2768.
    1. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol 2014;10:24–36.
    1. Son JS, Zhao L, Chen Y, Chen K, Chae SA, de Avila JM, et al. Maternal exercise via exerkine apelin enhances brown adipogenesis and prevents metabolic dysfunction in offspring mice. Sci Adv 2020;6:eaaz0359
    1. Silvester AJ, Aseer KR, Yun JW. Dietary polyphenols and their roles in fat browning. J Nutr Biochem 2019;64:1–12.
    1. Kulterer OC, Niederstaetter L, Herz CT, Haug AR, Bileck A, Pils D, et al. The presence of active brown adipose tissue determines cold-induced energy expenditure and oxylipin profiles in humans. J Clin Endocrinol Metab 2020;105:dgaa183
    1. Zoico E, Rubele S, De Caro A, Nori N, Mazzali G, Fantin F, et al. Brown and beige adipose tissue and aging. Front Endocrinol (Lausanne) 2019;10:368
    1. Wen J, Bo T, Zhang X, Wang Z, Wang D. Thermo-TRPs and gut microbiota are involved in thermogenesis and energy metabolism during low temperature exposure of obese mice. J Exp Biol 2020;223(Pt 11):jeb218974
    1. Hanssen MJ, Hoeks J, Brans B, van der Lans AA, Schaart G, van den Driessche JJ, et al. Short-term cold acclimation improves insulin sensitivity in patients with type 2 diabetes mellitus. Nat Med 2015;21:863–865.
    1. Cypess AM, Weiner LS, Roberts-Toler C, Franquet Elía E, Kessler SH, Kahn PA, et al. Activation of human brown adipose tissue by a β3-adrenergic receptor agonist. Cell Metab 2015;21:33–38.
    1. Baskin AS, Linderman JD, Brychta RJ, McGehee S, Anflick-Chames E, Cero C, et al. Regulation of human adipose tissue activation, gallbladder size, and bile acid metabolism by a β3-adrenergic receptor agonist. Diabetes 2018;67:2113–2125.
    1. Chen KY, Brychta RJ, Abdul Sater Z, Cassimatis TM, Cero C, Fletcher LA, et al. Opportunities and challenges in the therapeutic activation of human energy expenditure and thermogenesis to manage obesity. J Biol Chem 2020;295:1926–1942.
    1. O'Mara AE, Johnson JW, Linderman JD, Brychta RJ, Mc-Gehee S, Fletcher LA, et al. Chronic mirabegron treatment increases human brown fat, HDL cholesterol, and insulin sensitivity? J Clin Invest 2020;130:2209–2219.
    1. Kolettis TM, Kolettis MT. Winter swimming: healthy or hazardous? Evidence and hypotheses. Med Hypotheses 2003;61:654–656.
    1. Hermanussen M, Jensen F, Hirsch N, Friedel K, Kröger B, Lang R, et al. Acute and chronic effects of winter swimming on LH, FSH, prolactin, growth hormone, TSH, cortisol, serum glucose and insulin. Arctic Med Res 1995;54:45–51.
    1. Gibas-Dorna M, Checinska Z, Korek E, Kupsz J, Sowinska A, Wojciechowska M, et al. Variations in leptin and insulin levels within one swimming season in non-obese female cold water swimmers. Scand J Clin Lab Invest 2016;76:486–491.
    1. Lubkowska A, Dolegowska B, Szygula Z, Bryczkowska I, Stanczyk-Dunaj M, Salata D, et al. Winter-swimming as a building-up body resistance factor inducing adaptive changes in the oxidant/antioxidant status. Scand J Clin Lab Invest 2013;73:315–325.
    1. Manolis AS, Manolis SA, Manolis AA, Manolis TA, Apostolaki N, Melita H. Winter swimming: body hardening and cardiorespiratory protection via sustainable acclimation. Curr Sports Med Rep 2019;18:401–415.
    1. Ootsuka Y, McAllen RM. Comparison between two rat sympathetic pathways activated in cold defense. Am J Physiol Regul Integr Comp Physiol 2006;291:R589–R595.
    1. Rebello CJ, Greenway FL, Lau FH, Lin Y, Stephens JM, Johnson WD, et al. Naringenin promotes thermogenic gene expression in human white adipose tissue. Obesity (Silver Spring) 2019;27:103–111.
    1. Hubbard BP, Sinclair DA. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci 2014;35:146–154.
    1. Kim MY, Lim JH, Youn HH, Hong YA, Yang KS, Park HS, et al. Resveratrol prevents renal lipotoxicity and inhibits mesangial cell glucotoxicity in a manner dependent on the AMPK-SIRT1-PGC1α axis in db/db mice. Diabetologia 2013;56:204–217.
    1. Shixian Q, VanCrey B, Shi J, Kakuda Y, Jiang Y. Green tea extract thermogenesis-induced weight loss by epigallocatechin gallate inhibition of catechol-O-methyltransferase. J Med Food 2006;9:451–458.
    1. Cui H, Kong Y, Zhang H. Oxidative stress, mitochondrial dysfunction, and aging. J Signal Transduct 2012;2012:646354
    1. Lynes MD, Leiria LO, Lundh M, Bartelt A, Shamsi F, Huang TL, et al. The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue. Nat Med 2017;23:631–637.
    1. Leiria LO, Wang CH, Lynes MD, Yang K, Shamsi F, Sato M, et al. 12-Lipoxygenase regulates cold adaptation and glucose metabolism by producing the omega-3 lipid 12-hepe from brown fat. Cell Metab 2019;30:768–783.
    1. Ahmad TR, Haeusler RA. Bile acids in glucose metabolism and insulin signalling - mechanisms and research needs. Nat Rev Endocrinol 2019;15:701–712.
    1. Zietak M, Kozak LP. Bile acids induce uncoupling protein 1-dependent thermogenesis and stimulate energy expenditure at thermoneutrality in mice. Am J Physiol Endocrinol Metab 2016;310:E346–E354.
    1. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 2006;439:484–489.
    1. Kim KH, Choi S, Zhou Y, Kim EY, Lee JM, Saha PK, et al. Hepatic FXR/SHP axis modulates systemic glucose and fatty acid homeostasis in aged mice. Hepatology 2017;66:498–509.
    1. Strong R, Miller RA, Antebi A, Astle CM, Bogue M, Denzel MS, et al. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α-glucosidase inhibitor or a Nrf2-inducer. Aging Cell 2016;15:872–884.
    1. Amador-Noguez D, Dean A, Huang W, Setchell K, Moore D, Darlington G. Alterations in xenobiotic metabolism in the long-lived little mice. Aging Cell 2007;6:453–470.
    1. Cheng X, Maher J, Lu H, Klaassen CD. Endocrine regulation of gender-divergent mouse organic anion-transporting polypeptide (Oatp) expression. Mol Pharmacol 2006;70:1291–1297.
    1. Bárcena C, Quirós PM, Durand S, Mayoral P, Rodríguez F, Caravia XM, et al. Methionine restriction extends lifespan in progeroid mice and alters lipid and bile acid metabolism. Cell Rep 2018;24:2392–2403.
    1. Senn JR, Maushart CI, Gashi G, Michel R, Lalive d'Epinay M, Vogt R, et al. Outdoor temperature influences cold induced thermogenesis in humans. Front Physiol 2018;9:1184
    1. Folgueira C, Torres-Leal FL, Beiroa D, Pena-León V, Da Silva Lima N, Milbank E, et al. Oral pharmacological activation of hypothalamic guanylate cyclase 2C receptor stimulates brown fat thermogenesis to reduce body weight. Neuroendocrinology. 2020 [doi: 10.1159/000505972]
      [Epub].
    1. Alexopoulos SJ, Chen SY, Brandon AE, Salamoun JM, Byrne FL, Garcia CJ, et al. Mitochondrial uncoupler BAM15 reverses diet-induced obesity and insulin resistance in mice. Nat Commun 2020;11:2397
    1. Mahú I, Barateiro A, Rial-Pensado E, Martinéz-Sánchez N, Vaz SH, Cal PMSD, et al. Brain-sparing sympathofacilitators mitigate obesity without adverse cardiovascular effects. Cell Metab 2020;31:1120–1135.e7.
    1. Bernier M, Mitchell SJ, Wahl D, Diaz A, Singh A, Seo W, et al. Disulfiram treatment normalizes body weight in obese mice. Cell Metab. 2020 [doi: 10.1016/j.cmet.2020.04.019]
      [Epub].
    1. Tajima K, Ikeda K, Tanabe Y, Thomson EA, Yoneshiro T, Oguri Y, et al. Wireless optogenetics protects against obesity via stimulation of non-canonical fat thermogenesis. Nat Commun 2020;11:1730

Metrics
Share
PERMALINK