Next Article in Journal
Intermediate-Level Diet Quality of Brazilian Paralympic Athletes Based on National and International Indexes
Next Article in Special Issue
Males Have a Higher Energy Expenditure than Females during Squat Training
Previous Article in Journal
Fructose Metabolism and Metabolic Dysfunction in Adolescents and Young Adults
Previous Article in Special Issue
Influence of 12-Week Concurrent Training on Exosome Cargo and Its Relationship with Cardiometabolic Health Parameters in Men with Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Exercise Is Medicine: How Do We Implement It?

by
Aliya Khasanova
1,2 and
Tara M. Henagan
1,2,*
1
Department of Family Medicine, Baton Rouge General Family Health Center, Baton Rouge, LA 70806, USA
2
Department of Family Medicine, Baton Rouge General Hospital, Baton Rouge, LA 70808, USA
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(14), 3164; https://doi.org/10.3390/nu15143164
Submission received: 26 June 2023 / Revised: 8 July 2023 / Accepted: 12 July 2023 / Published: 17 July 2023

Abstract

:
Exercise is well known to have beneficial effects on various disease states. In this paper, we broadly describe the fundamental concepts that are shared among various disease states, including obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), heart failure (HF), cancer, and psychological well-being, and the beneficial effects of exercise training within these concepts. We highlight issues involved in implementing exercise recommendations and describe the potential impacts and challenges to medical professionals and patients. Problems are identified and discussed with respect to the future roles of professionals in the current built environment with its limited infrastructure to support current physical activity recommendations.

1. Exercise and Energy Expenditure

Exercise has been shown to improve outcomes in a variety of disease states, as well as in preventing disease [1]. The effects of exercise for the prevention and treatment of disease may be due to its effects on energy regulation [1]. It is well known that energy regulation is dependent on the intensity of exercise training [2], with lower-intensity endurance training activating aerobic respiratory pathways and high-intensity aerobic training and weight training activating more anaerobic respiratory pathways [3,4]. For example, marathon training increases beta oxidation of fatty acids in addition to the glycolytic catabolism of carbohydrates stored in the form of glycogen as well as those carbohydrates ingested during the training session [5,6]. Conversely, high-intensity interval training (HIIT) activates creatine kinase to break down glycogen quickly and increases glycolysis for glucose catabolism, sparing fatty acids [4,6]. Although aerobic exercise is known to increase fatty acid oxidation during an exercise bout [7], anaerobic exercise has been shown to increase the capacity of fatty acid oxidation in the post-exercise interval as well as throughout the day by increasing the basal metabolic rate and muscle mass [8,9]. Increased muscle mass contributes to increases in the thermic effect of exercise as well as increases in the basal metabolic rate [8,9]. Thus, anaerobic exercise may also lead to more long-term fatty acid oxidation outside of the actual exercise bout. Whether aerobic or anaerobic in nature, exercise training increases overall total energy expenditure. Although energy balance in humans is much more complex than simply considering energy intake vs. energy expended, in theory and in the most simplistic terms, exercise training in the absence of changes in energy intake leads to negative energy balance and weight loss.

2. Exercise and Obesity and T2D

One of the largest crises that the United States currently faces is the obesity epidemic, which is closely related to the increased incidence and prevalence of CVD and T2D. In the most simplistic of theories and terms, obesity is a state of positive energy balance, where energy intake exceeds energy expenditure. Given the beneficial effects of both aerobic and anaerobic exercise on energy metabolism, the potential of exercise training to prevent and treat obesity via its ability to increase fatty acid oxidation and energy expenditure is clear [7,10]. In turn, weight loss, even as little as 5% of one’s body weight, is known to improve insulin sensitivity and T2D [11]. Additionally, skeletal muscle contraction itself is known to activate GLUT4 receptors to increase glucose uptake into skeletal muscle, independent of insulin [12]. Thus, unique to exercise as a form of medicine in the treatment of T2D, exercise itself, without changes in body weight, can improve insulin sensitivity and ameliorate T2D. Obesity is a state that is also defined by chronic inflammation [13]. In fact, it has been shown that chronic inflammation may be one of the causative factors in determining an obese state or in maintaining obesity. Chronic inflammation is also linked to insulin resistance and T2D, and alleviating chronic inflammation has been shown to improve insulin sensitivity and reverse T2D [14]. Exercise training, both aerobic and anaerobic training, is known to acutely increase inflammation, largely due to microenvironmental insults that occur during an exercise bout [15]. However, in the recovery period following exercise, this acute increase in inflammatory markers, such as cytokines, leads to recruitment and activation of anti-inflammatory cells and pathways [16,17]. Activation of these anti-inflammatory pathways leads to long-term downregulation of chronic inflammation in response to repeated exercise bouts [17,18,19]. Given the long-term exercise-induced reductions in chronic inflammation, exercise training may play a large role in preventing and treating diseases associated with chronic inflammation.

3. Exercise and CVD and HF

CVD is marked by increased levels of inflammation and atherosclerosis and is also prevented and treated with exercise [20,21]. Atherosclerosis is marked by decreased vascular compliance, which plays a role in hypertension and CVD [22,23]. Aortic stiffness or lack of compliance has been inversely correlated with low density lipoprotein (LDL) levels and positively correlated with the amount of energy expended during physical activity [22], and exercise training itself has been shown to improve arterial compliance [23]. Similar to its effects in T2D, exercise also increases fatty acid oxidation and decreases LDL cholesterol, the substrate known to largely contribute to decreased arterial compliance, atherosclerosis, and increased CVD risk [7,24,25,26]. As previously discussed, beyond its ability to alter fatty acid and cholesterol metabolism, exercise training also decreases chronic inflammation. Chronic inflammation is a contributing event in the onset and progression of atherosclerosis and CVD [21]. Collectively, exercise training improves vascular compliance and vascular health and decreases CVD risk [27,28]. In addition to the effects of chronic inflammation in the pathogenesis of CVD, studies have also found that inflammation contributes to HF. Indeed, inflammatory markers have been positively correlated with the progression, prognosis, and severity of HF [29], and physical activity levels are inversely related to HF risk and incidence [30,31]. The benefits of exercise training for HF risk reduction and prevention may be attributed to its effects on cardiac muscle and remodeling and also on chronic inflammation [32,33].

4. Exercise and Cancer

Increased physical activity levels and exercise training have been linked to cancer prevention, with longer time spent in sedentary behaviors being associated with cancer risk and with increased physical activity levels and exercise positively impacting breast, colon, and ovarian cancers and multiple myeloma [34]. Similar to its effects in the prevention and treatment of other disease states, exercise training may act to prevent cancer by modulating chronic inflammation, a contributing factor in cancer onset and progression [35]. Exercise may also regulate cellular growth and cancer by altering levels of estrogen and growth factors that are involved in uncontrolled cellular proliferation [36]. Interestingly, obesity and insulin resistance are also risk factors that appear to be involved in cancer onset and that impact cancer risk [37,38,39]. Metabolic adaptations that occur during obesity, including increased levels of fatty acids and blood glucose levels, increase the available substrates for cancer cell growth and metabolism [40,41]. The increased energy expended during and following exercise alters the microenvironment by increasing fatty acid and glucose uptake and utilization by skeletal muscle and other peripheral tissues, limiting available resources for cancer cells and inhibiting tumor growth [42].

5. Psychological Benefits of Exercise

In addition to its beneficial effects on energy metabolism, exercise training has been shown to improve psychological well-being [43]. For example, exercise training has been shown to improve depression and mental health with exercise-induced psychological improvements similar to those seen in response to antidepressant therapies [44]. Serum concentrations of endogenous opioids, in particular β-endorphin and β-lipotrophin, are elevated during exercise or training [45]. Elevated β-endorphins during exercise have been linked to several psychological and physiological changes, including mood state changes and exercise-induced euphoria, decreases in pain perception, and positive alterations in stress response hormones, including cortisol [46,47,48]. Exercise-induced improvements in depression have also been associated with exercise-induced improvements in chronic inflammation, as depression is associated with chronic inflammation, and exercise improves depression and decreases chronic inflammation [49,50]. Physical activity levels have also been associated with cognitive functioning and memory, with individuals with lower physical activity levels having a higher risk for developing cognitive impairment and dementia later in life [51]. Interestingly, aging alone is associated with increased levels of chronic inflammation [52]. In individuals with cognitive impairment, exercise training has been shown to increase cognitive function [53], and in individuals with Alzheimer’s disease, it increases memory performance [54,55]. Indeed, both aerobic and anaerobic exercise training have been shown to improve memory, and these improvements have been associated with decreased inflammation [56,57,58].

6. Exercise Protects Future Generations

Exercise training not only has the ability to alter the course of disease in a specific individual but also has the ability to prevent disease in future generations. Exercise not only improves outcomes in obesity, T2D, CVD, etc., but also alters the epigenome. Exercise impacts HDAC activity [59], histone acetylation [60], histone methylation [61], histone phosphorylation [62], DNA methylation [63,64], nucleosome positioning [64,65], and, it has been postulated, lactylation [66]. Furthermore, exercise-induced epigenetic alterations have been shown to be inherited by offspring [67]. Interestingly, exercise during pregnancy and paternal exercise in the preconception period both improve glucose tolerance in offspring [68,69], and exercise training attenuates insulin resistance in offspring fed a high-fat diet [70]. Additionally, the effects of maternal exercise may abolish any detrimental metabolic effects of obesity on offspring via epigenetic mechanisms [44]. While it is clear that exercise training affects epigenetic modifications and these modifications are heritable and improve obesity- and T2DM-related outcomes [59,60,61,62,63,64,65,66,67,68,69,70], studies on the potential heritable effects of exercise on CVD, stroke, cancer, etc., are lacking. However, given the pleotropic effects of exercise in preventing and improving T2D and obesity and its ability to alter the epigenome [10,12,59,60,61,62,63,64,65,66,67,68,69,70], it is possible that the effects of maternal and paternal exercise are far-reaching and affect offspring outcomes with respect to other chronic disease states.

7. Clinical Recommendations for Implementing Exercise and Barriers

Current clinical recommendations in the Unites States are stratified by age [71]. It is recommended that US adults participate in 150–300 min of moderate-intensity or 75–150 min of vigorous-intensity aerobic physical activity per week, with this activity being spread out throughout the week [71]. In addition, more recent recommendations have acknowledged the health benefits of anaerobic training in the form of resistance training and incorporated the guideline that US adults should participate in muscle-strengthening activities of moderate or greater intensity that involve all major muscle groups on two or more days a week [71]. For most clinic patients, incorporating physical activity and exercise guidelines into their everyday lives is difficult. There are many barriers to doing so in many parts of the US, including access to a safe place to perform exercise training, access to weight training equipment or gyms, cost of memberships at facilities where equipment is available, travel distance and time to and from facilities, working knowledge of how to start and maintain an exercise routine to continue to reap the health benefits of exercise training, how to optimize recovery from training, etc. [72,73]. Although medical doctors are aware of the Physical Activity Guidelines for Americans and many regularly counsel their patients on such guidelines, most medical doctors are not trained or knowledgeable regarding how to create an exercise plan, especially weight training plans based on the various goals of this exercise modality, how to counsel patients on post-exercise recovery periods, how to counsel patients on pre- and post-exercise nutrition, what constitutes “moderate” and “vigorous” exercise, the best exercise modalities for individuals given physical limitations, etc. [74].

8. Conclusions

It is clear that exercise training, both aerobic and anaerobic, has far-reaching beneficial effects on health and disease prevention and treatment. Clinical guidelines are available for the prescription of physical activity for Americans, yet these guidelines are lacking in sufficient detail for specific recommendations for various populations using various modalities of exercise due to the limitations of current scientific research that has investigated the specific benefits of exercise modality, intensity, duration, etc., with respect to disease states in different age groups and populations to determine the best approach to using exercise training for disease prevention and treatment. In addition, throughout most of the US, where chronic diseases that can be easily prevented or treated with exercise cost $4.1 trillion in healthcare [71,75], the infrastructure to support more healthy physical activity is lacking. An easily accomplished first step to building this infrastructure is to utilize a more comprehensive approach to healthcare. One can easily envision an approach that can be adopted where general practitioners, such as family medicine physicians, are at the forefront of coordinating and helping patients implement dietary and exercise interventions for optimal health. In order for physicians to do this, support and buy-in from local and state agencies is required. Something as simple as building more sidewalks in neighborhoods and along streets, building and maintaining safe parks and recreational facilities that are “free” to the public, integrating biking paths that are safe to travel along streets, even incorporating gyms in local physician practices whose membership fees are covered by health insurance are some of the possible solutions that can start to provide the care needed to protect and improve the health of all Americans (Figure 1).

Author Contributions

Conceptualization, T.M.H.; writing—original draft preparation, T.M.H. and A.K.; writing—review and editing, T.M.H. and A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Martin, M.; Krystof, S.; Jiri, R.; Martina, D.; Renata, V.; Ondrej, M.; Stepan, S.; Vladimir, T. Modulation of Energy Intake and Expenditure Due to Habitual Physical Exercise. Curr. Pharm. Des. 2016, 22, 3681–3699. [Google Scholar] [CrossRef] [PubMed]
  2. Broskey, N.T.; Martin, C.K.; Burton, J.H.; Church, T.S.; Ravussin, E.; Redman, L.M. Effect of Aerobic Exercise-induced Weight Loss on the Components of Daily Energy Expenditure. Med. Sci. Sport. Exerc. 2021, 53, 2164–2172. [Google Scholar] [CrossRef] [PubMed]
  3. Mang, Z.A.; Ducharme, J.B.; Mermier, C.; Kravitz, L.; Magalhaes, F.D.C.; Amorim, F. Aerobic Adaptations to Resistance Training: The Role of Time under Tension. Int. J. Sport. Med. 2022, 43, 829–839. [Google Scholar] [CrossRef]
  4. Jeppesen, J.; Kiens, B. Regulation and limitations to fatty acid oxidation during exercise. J. Physiol. 2012, 590, 1059–1068. [Google Scholar] [CrossRef] [PubMed]
  5. Mika, A.; Macaluso, F.; Barone, R.; Di Felice, V.; Sledzinski, T. Effect of Exercise on Fatty Acid Metabolism and Adipokine Secretion in Adipose Tissue. Front. Physiol. 2019, 10, 26. [Google Scholar] [CrossRef] [Green Version]
  6. Achten, J.; Gleeson, M.; Jeukendrup, A.E. Determination of the exercise intensity that elicits maximal fat oxidation. Med. Sci. Sport. Exerc. 2002, 34, 92–97. [Google Scholar] [CrossRef] [PubMed]
  7. Horowitz, J.F.; Klein, S. Lipid metabolism during endurance exercise. Am. J. Clin. Nutr. 2000, 72, 558S–563S. [Google Scholar] [CrossRef] [Green Version]
  8. Hunter, G.R.; Weinsier, R.L.; Bamman, M.M.; Larson, D.E. A role for high intensity exercise on energy balance and weight control. Int. J. Obes. Relat. Metab. Disord. 1998, 22, 489–493. [Google Scholar] [CrossRef] [Green Version]
  9. Osterberg, K.L.; Melby, C.L. Effect of acute resistance exercise on postexercise oxygen consumption and resting metabolic rate in young women. Int. J. Sport Nutr. Exerc. Metab. 2000, 10, 71–81. [Google Scholar] [CrossRef]
  10. Petridou, A.; Siopi, A.; Mougios, V. Exercise in the management of obesity. Metabolism 2019, 92, 163–169. [Google Scholar] [CrossRef] [Green Version]
  11. Ryan, D.H.; Yockey, S.R. Weight Loss and Improvement in Comorbidity: Differences at 5%, 10%, 15%, and Over. Curr. Obes. Rep. 2017, 6, 187–194. [Google Scholar] [CrossRef] [PubMed]
  12. Kennedy, J.W.; Hirshman, M.F.; Gervino, E.V.; Ocel, J.V.; A Forse, R.; Hoenig, S.J.; Aronson, D.; Goodyear, L.J.; Horton, E.S. Acute exercise induces GLUT4 translocation in skeletal muscle of normal human subjects and subjects with type 2 diabetes. Diabetes 1999, 48, 1192–1197. [Google Scholar] [CrossRef] [PubMed]
  13. Khanna, D.; Khanna, S.; Khanna, P.; Kahar, P.; Patel, B.M. Obesity: A Chronic Low-Grade Inflammation and Its Markers. Cureus 2022, 14, e22711. [Google Scholar] [CrossRef]
  14. Wu, H.; Ballantyne, C.M. Metabolic Inflammation and Insulin Resistance in Obesity. Circ. Res. 2020, 126, 1549–1564. [Google Scholar] [CrossRef] [PubMed]
  15. Peake, J.M.; Neubauer, O.; Della Gatta, P.A.; Nosaka, K. Muscle damage and inflammation during recovery from exercise. J. Appl. Physiol. 2017, 122, 559–570. [Google Scholar] [CrossRef]
  16. Pedersen, B.K. Muscles and their myokines. J. Exp. Biol. 2011, 214 Pt 2, 337–346. [Google Scholar] [CrossRef] [Green Version]
  17. Henagan, T.M.; Phillips, M.D.; Cheek, D.J.; Kirk, K.M.; Barbee, J.J.; Stewart, L.K. The melanocortin 3 receptor: A novel mediator of exercise-induced inflammation reduction in postmenopausal women? J. Aging Res. 2011, 2011, 512593. [Google Scholar] [CrossRef] [Green Version]
  18. Henagan, T.M.; Forney, L.; Dietrich, M.A.; Harrell, B.R.; Stewart, L.K. Melanocortin receptor expression is associated with reduced CRP in response to resistance training. J. Appl. Physiol. 2012, 113, 393–400. [Google Scholar] [CrossRef] [Green Version]
  19. Scott, V.; O’Brien, K.; Boeneke, C.; Stewart, L.; Forney, L.; Henagan, T. Exercise- and Kefir-induced Internalization of the Anti-inflammatory Melanocortin 3 Receptor in Monocytes. FASEB J. 2015, 29, 677.29. [Google Scholar] [CrossRef]
  20. Dempsey, P.C.; Rowlands, A.V.; Strain, T.; Zaccardi, F.; Dawkins, N.; Razieh, C.; Davies, M.J.; Khunti, K.K.; Edwardson, C.L.; Wijndaele, K.; et al. Physical activity volume, intensity, and incident cardiovascular disease. Eur. Heart J. 2022, 43, 4789–4800. [Google Scholar] [CrossRef]
  21. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  22. Kupari, M.; Hekali, P.; Keto, P.; Poutanen, V.P.; Tikkanen, M.J.; Standerstkjöld-Nordenstam, C.G. Relation of aortic stiffness to factors modifying the risk of atherosclerosis in healthy people. Arter. Thromb. A J. Vasc. Biol. 1994, 14, 386–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Cameron, J.D.; Dart, A.M. Exercise training increases total systemic arterial compliance in humans. Am. J. Physiol. Circ. Physiol. 1994, 266 Pt 2, H693–H701. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, W.D.; Wang, Y.; Wei, J.X. [Effects of MICT/HIIT on the ultrastructure of myocardium and soleus in rats with high-fat diet and its mechanisms]. Chin. J. Appl. Physiol. 2022, 38, 708–713. [Google Scholar]
  25. Halverstadt, A.; Phares, D.A.; Wilund, K.R.; Goldberg, A.P.; Hagberg, J.M. Endurance exercise training raises high-density lipoprotein cholesterol and lowers small low-density lipoprotein and very low-density lipoprotein independent of body fat phenotypes in older men and women. Metabolism 2007, 56, 444–450. [Google Scholar] [CrossRef]
  26. Cromwell, W.C.; Otvos, J.D. Low-density lipoprotein particle number and risk for cardiovascular disease. Curr. Atheroscler. Rep. 2004, 6, 381–387. [Google Scholar] [CrossRef]
  27. Laurent, P.; Marenco, P.; Castagna, O.; Smulyan, H.; Blacher, J.; Safar, M.E. Differences in central systolic blood pressure and aortic stiffness between aerobically trained and sedentary individuals. J. Am. Soc. Hypertens. 2011, 5, 85–93. [Google Scholar] [CrossRef] [PubMed]
  28. Nualnim, N.; Parkhurst, K.; Dhindsa, M.; Tarumi, T.; Vavrek, J.; Tanaka, H. Effects of Swimming Training on Blood Pressure and Vascular Function in Adults >50 Years of Age. Am. J. Cardiol. 2012, 109, 1005–1010. [Google Scholar] [CrossRef] [Green Version]
  29. Dick, S.A.; Epelman, S. Chronic Heart Failure and Inflammation: What Do We Really Know? Circ. Res. 2016, 119, 159–176. [Google Scholar] [CrossRef]
  30. Kraigher-Krainer, E.; Lyass, A.; Massaro, J.M.; Lee, D.S.; Ho, J.E.; Levy, D.; Kannel, W.B.; Vasan, R.S. Association of physical activity and heart failure with preserved vs. reduced ejection fraction in the elderly: The Framingham Heart Study. Eur. J. Heart Fail. 2013, 15, 742–746. [Google Scholar] [CrossRef] [Green Version]
  31. Pandey, A.; Garg, S.; Khunger, M.; Darden, D.; Ayers, C.; Kumbhani, D.J.; Mayo, H.G.; de Lemos, J.A.; Berry, J.D. Dose-Response Relationship Between Physical Activity and Risk of Heart Failure: A Meta-Analysis. Circulation 2015, 132, 1786–1794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Brinker, S.K.; Pandey, A.; Ayers, C.R.; Barlow, C.E.; DeFina, L.F.; Willis, B.L.; Radford, N.B.; Farzaneh-Far, R.; de Lemos, J.A.; Drazner, M.H.; et al. Association of cardiorespiratory fitness with left ventricular remodeling and diastolic function: The Cooper Center Longitudinal Study. JACC Heart Fail. 2014, 2, 238–246. [Google Scholar] [CrossRef]
  33. Nunes, R.B.; Alves, J.P.; Kessler, L.P.; Lago, P.D. Aerobic exercise improves the inflammatory profile correlated with cardiac remodeling and function in chronic heart failure rats. Clinics 2013, 68, 876–882. [Google Scholar] [CrossRef]
  34. Patel, A.V.; Hildebrand, J.S.; Campbell, P.T.; Teras, L.R.; Craft, L.L.; McCullough, M.L.; Gapstur, S.M. Leisure-Time Spent Sitting and Site-Specific Cancer Incidence in a Large U.S. Cohort. Cancer Epidemiol. Biomark. Prev. 2015, 24, 1350–1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Koelwyn, G.J.; Wennerberg, E.; Demaria, S.; Jones, L.W. Exercise in Regulation of Inflammation-Immune Axis Function in Cancer Initiation and Progression. Oncology 2015, 29, 214800. [Google Scholar] [PubMed]
  36. Wang, Q.; Zhou, W. Roles and molecular mechanisms of physical exercise in cancer prevention and treatment. J. Sport Health Sci. 2021, 10, 201–210. [Google Scholar] [CrossRef] [PubMed]
  37. Luque, R.M.; López-Sánchez, L.M.; Villa-Osaba, A.; Luque, I.M.; Santos-Romero, A.L.; Yubero-Serrano, E.M.; Cara-García, M.; Álvarez-Benito, M.; López-Miranda, J.; Gahete, M.D.; et al. Breast cancer is associated to impaired glucose/insulin homeostasis in premenopausal obese/overweight patients. Oncotarget 2017, 8, 81462–81474. [Google Scholar] [CrossRef] [Green Version]
  38. Yoshikawa, T.; Noguchi, Y.; Doi, C.; Makino, T.; Nomura, K. Insulin resistance in patients with cancer: Relationships with tumor site, tumor stage, body-weight loss, acute-phase response, and energy expenditure. Nutrition 2001, 17, 590–593. [Google Scholar] [CrossRef]
  39. Schoen, R.E.; Tangen, C.M.; Kuller, L.H.; Burke, G.L.; Cushman, M.; Tracy, R.P.; Dobs, A.; Savage, P.J. Increased Blood Glucose and Insulin, Body Size, and Incident Colorectal Cancer. Gynecol. Oncol. 1999, 91, 1147–1154. [Google Scholar] [CrossRef] [Green Version]
  40. Rapp, K.; Schroeder, J.; Klenk, J.; Ulmer, H.; Concin, H.; Diem, G.; Oberaigner, W.; Weiland, S.K. Fasting blood glucose and cancer risk in a cohort of more than 140,000 adults in Austria. Diabetologia 2006, 49, 945–952. [Google Scholar] [CrossRef] [Green Version]
  41. Warburg, O. The Metabolism of Carcinoma Cells. J. Cancer Res. 1925, 9, 148–163. [Google Scholar] [CrossRef] [Green Version]
  42. Moreira, V.M.; da Silva Franco, C.C.; Prates, K.V.; Gomes, R.M.; de Moraes, A.M.P.; Ribeiro, T.A.; Martins, I.P.; Previate, C.; Pavanello, A.; Matiusso, C.C.I.; et al. Aerobic Exercise Training Attenuates Tumor Growth and Reduces Insulin Secretion in Walker 256 Tumor-Bearing Rats. Front. Physiol. 2018, 9, 465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Balchin, R.; Linde, J.; Blackhurst, D.; Rauch, H.L.; Schönbächler, G. Sweating away depression? The impact of intensive exercise on depression. J. Affect. Disord. 2016, 200, 218–221. [Google Scholar] [CrossRef]
  44. Laker, R.C.; Altıntaş, A.; Lillard, T.S.; Zhang, M.; Connelly, J.J.; Sabik, O.L.; Onengut, S.; Rich, S.S.; Farber, C.R.; Barrès, R.; et al. Exercise during pregnancy mitigates negative effects of parental obesity on metabolic function in adult mouse offspring. J. Appl. Physiol. 2021, 130, 605–616. [Google Scholar] [CrossRef] [PubMed]
  45. Goldfarb, A.H.; Jamurtas, A.Z. Beta-endorphin response to exercise. An update. Sport. Med. 1997, 24, 8–16. [Google Scholar] [CrossRef] [PubMed]
  46. Netz, Y. Is the Comparison between Exercise and Pharmacologic Treatment of Depression in the Clinical Practice Guideline of the American College of Physicians Evidence-Based? Front. Pharmacol 2017, 8, 257. [Google Scholar] [CrossRef] [Green Version]
  47. Heyman, E.; Gamelin, F.-X.; Goekint, M.; Piscitelli, F.; Roelands, B.; Leclair, E.; Di Marzo, V.; Meeusen, R. Intense exercise increases circulating endocannabinoid and BDNF levels in humans—Possible implications for reward and depression. Psychoneuroendocrinology 2012, 37, 844–851. [Google Scholar] [CrossRef]
  48. Caplin, A.; Chen, F.; Beauchamp, M.; Puterman, E. The effects of exercise intensity on the cortisol response to a subsequent acute psychosocial stressor. Psychoneuroendocrinology 2021, 131, 105336. [Google Scholar] [CrossRef]
  49. Paolucci, E.M.; Loukov, D.; Bowdish, D.M.; Heisz, J.J. Exercise reduces depression and inflammation but intensity matters. Biol. Psychol. 2018, 133, 79–84. [Google Scholar] [CrossRef]
  50. Moussavi, S.; Chatterji, S.; Verdes, E.; Tandon, A.; Patel, V.; Ustun, B. Depression, chronic diseases, and decrements in health: Results from the World Health Surveys. Lancet 2007, 370, 851–858. [Google Scholar] [CrossRef]
  51. Buchman, A.; Boyle, P.; Yu, L.; Shah, R.; Wilson, R.; Bennett, D. Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology 2012, 78, 1323–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Bruunsgaard, H.; Pedersen, A.; Schroll, M.; Skinhøj, P.; Pedersen, B. TNF-α, leptin, and lymphocyte function in human aging. Life Sci. 2000, 67, 2721–2731. [Google Scholar] [CrossRef]
  53. Hong, S.-G.; Kim, J.-H.; Jun, T.-W. Effects of 12-Week Resistance Exercise on Electroencephalogram Patterns and Cognitive Function in the Elderly With Mild Cognitive Impairment: A Randomized Controlled Trial. Clin. J. Sport Med. 2018, 28, 500–508. [Google Scholar] [CrossRef] [PubMed]
  54. Morris, J.K.; Vidoni, E.D.; Johnson, D.K.; Van Sciver, A.; Mahnken, J.D.; Honea, R.A.; Wilkins, H.M.; Brooks, W.M.; Billinger, S.A.; Swerdlow, R.H.; et al. Aerobic exercise for Alzheimer's disease: A randomized controlled pilot trial. PLoS ONE 2017, 12, e0170547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Öhman, H.; Savikko, N.; Strandberg, T.E.; Kautiainen, H.; Raivio, M.M.; Laakkonen, M.-L.; Tilvis, R.; Pitkälä, K.H. Effects of Exercise on Cognition: The Finnish Alzheimer Disease Exercise Trial: A Randomized, Controlled Trial. J. Am. Geriatr. Soc. 2016, 64, 731–738. [Google Scholar] [CrossRef]
  56. De la Rosa, A.; Solana, E.; Corpas, R.; Bartrés-Faz, D.; Pallàs, M.; Vina, J.; Sanfeliu, C.; Gomez-Cabrera, M.C. Long-term exercise training improves memory in middle-aged men and modulates peripheral levels of BDNF and Cathepsin, B. Sci. Rep. 2019, 9, 3337. [Google Scholar] [CrossRef] [Green Version]
  57. Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al. Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022. [Google Scholar] [CrossRef]
  58. Fiatarone Singh, M.A.; Gates, N.; Saigal, N.; Wilson, G.C.; Meiklejohn, J.; Brodaty, H.; Wen, W.; Singh, N.; Baune, B.T.; Suo, C.; et al. The Study of Mental and Resistance Training (SMART) study—Resistance training and/or cognitive training in mild cognitive impairment: A randomized, double-blind, double-sham controlled trial. J. Am. Med. Dir. Assoc. 2014, 15, 873–880. [Google Scholar] [CrossRef]
  59. McGee, S.L.; Fairlie, E.; Garnham, A.P.; Hargreaves, M. Exercise-induced histone modifications in human skeletal muscle. J. Physiol. 2009, 587 Pt 24, 5951–5958. [Google Scholar] [CrossRef]
  60. Lim, C.; Shimizu, J.; Kawano, F.; Kim, H.J.; Kim, C.K. Adaptive responses of histone modifications to resistance exercise in human skeletal muscle. PLoS ONE 2020, 15, e0231321. [Google Scholar] [CrossRef] [Green Version]
  61. Shimizu, J.; Kawano, F. Exercise-induced histone H3 trimethylation at lysine 27 facilitates the adaptation of skeletal muscle to exercise in mice. J. Physiol. 2022, 600, 3331–3353. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, M.; Stepto, N.K.; Chibalin, A.V.; Fryer, L.G.D.; Carling, D.; Krook, A.; Hawley, J.A.; Zierath, J.R. Metabolic and mitogenic signal transduction in human skeletal muscle after intense cycling exercise. J. Physiol. 2003, 546, 327–335. [Google Scholar] [CrossRef] [PubMed]
  63. Barrès, R.; Yan, J.; Egan, B.; Treebak, J.T.; Rasmussen, M.; Fritz, T.; Caidahl, K.; Krook, A.; O'Gorman, D.J.; Zierath, J.R. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012, 15, 405–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bajpeyi, S.; Covington, J.D.; Taylor, E.M.; Stewart, L.K.; Galgani, J.E.; Henagan, T.M. Skeletal Muscle PGC1α −1 Nucleosome Position and −260 nt DNA Methylation Determine Exercise Response and Prevent Ectopic Lipid Accumulation in Men. Endocrinology 2017, 158, 2190–2199. [Google Scholar] [CrossRef] [PubMed]
  65. Devarshi, P.P.; Pereyra, A.S.; Ellis, J.M.; Henagan, T.M. A single bout of cycling exercise induces nucleosome repositioning in the skeletal muscle of lean and overweight/obese individuals. Diabetes Obes. Metab. 2022, 24, 21–33. [Google Scholar] [CrossRef]
  66. Seaborne, R.A.; Sharples, A.P. The Interplay Between Exercise Metabolism, Epigenetics, and Skeletal Muscle Remodeling. Exerc. Sport Sci. Rev. 2020, 48, 188–200. [Google Scholar] [CrossRef]
  67. Axsom, J.E.; Libonati, J.R. Impact of parental exercise on epigenetic modifications inherited by offspring: A systematic review. Physiol. Rep. 2019, 7, e14287. [Google Scholar] [CrossRef] [Green Version]
  68. Carter, L.G.; Qi, N.R.; DE Cabo, R.; Pearson, K.J. Maternal Exercise Improves Insulin Sensitivity in Mature Rat Offspring. Med. Sci. Sport. Exerc. 2013, 45, 832–840. [Google Scholar] [CrossRef] [Green Version]
  69. Stanford, K.I.; Rasmussen, M.; Baer, L.A.; Lehnig, A.C.; Rowland, L.A.; White, J.D.; So, K.; De Sousa-Coelho, A.L.; Hirshman, M.F.; Patti, M.-E.; et al. Paternal Exercise Improves Glucose Metabolism in Adult Offspring. Diabetes 2018, 67, 2530–2540. [Google Scholar] [CrossRef] [Green Version]
  70. Laker, R.C.; Lillard, T.S.; Okutsu, M.; Zhang, M.; Hoehn, K.L.; Connelly, J.J.; Yan, Z. Exercise Prevents Maternal High-Fat Diet–Induced Hypermethylation of the Pgc-1α Gene and Age-Dependent Metabolic Dysfunction in the Offspring. Diabetes 2014, 63, 1605–1611. [Google Scholar] [CrossRef] [Green Version]
  71. Piercy, K.L.; Troiano, R.P.; Ballard, R.M.; Carlson, S.A.; Fulton, J.E.; Galuska, D.A.; George, S.M.; Olson, R.D. The Physical Activity Guidelines for Americans. JAMA 2018, 320, 2020–2028. [Google Scholar] [CrossRef] [PubMed]
  72. Bantham, A.; Ross, S.E.T.; Sebastião, E.; Hall, G. Overcoming barriers to physical activity in underserved populations. Prog. Cardiovasc. Dis. 2021, 64, 64–71. [Google Scholar] [CrossRef] [PubMed]
  73. Herazo-Beltrán, Y.; Pinillos, Y.; Vidarte, J.; Crissien, E.; Suarez, D.; García, R. Predictors of perceived barriers to physical activity in the general adult population: A cross-sectional study. Braz. J. Phys. Ther. 2017, 21, 44–50. [Google Scholar] [CrossRef]
  74. Cardinal, B.J.; Park, E.A.; Kim, M.; Cardinal, M.K. If Exercise is Medicine, Where is Exercise in Medicine? Review of U.S. Medical Education Curricula for Physical Activity-Related Content. J. Phys. Act. Health 2015, 12, 1336–1343. [Google Scholar] [CrossRef] [PubMed]
  75. National Health Expenditure Data: Historical. Center for Medicare & Medicaid Services. 2021. Available online: https://www.cms.gov/research-statistics-data-and-systems/statistics-trends-and-reports/nationalhealthexpenddata/nationalhealthaccountshistorical (accessed on 11 July 2023).
Figure 1. Exercise has many beneficial effects, including but not limited to prevention of and improvement in psychological, cardiovascular, obesity and type 2 diabetes mellitus (T2DM), cancer, and epigenetics disease states and outcomes, respectively. Despite the known beneficial effects of exercise, many barriers still exist in implementing exercise and physical activity in clinical practice for disease prevention and treatment. These barriers and how to reduce them deserve more attention in providing holistic, evidence-based best practices in patient care.
Figure 1. Exercise has many beneficial effects, including but not limited to prevention of and improvement in psychological, cardiovascular, obesity and type 2 diabetes mellitus (T2DM), cancer, and epigenetics disease states and outcomes, respectively. Despite the known beneficial effects of exercise, many barriers still exist in implementing exercise and physical activity in clinical practice for disease prevention and treatment. These barriers and how to reduce them deserve more attention in providing holistic, evidence-based best practices in patient care.
Nutrients 15 03164 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Khasanova, A.; Henagan, T.M. Exercise Is Medicine: How Do We Implement It? Nutrients 2023, 15, 3164. https://doi.org/10.3390/nu15143164

AMA Style

Khasanova A, Henagan TM. Exercise Is Medicine: How Do We Implement It? Nutrients. 2023; 15(14):3164. https://doi.org/10.3390/nu15143164

Chicago/Turabian Style

Khasanova, Aliya, and Tara M. Henagan. 2023. "Exercise Is Medicine: How Do We Implement It?" Nutrients 15, no. 14: 3164. https://doi.org/10.3390/nu15143164

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop