EXOSOMES (26).png
 

White Paper by Kritika Pathak

Reviewed by Ben Kronz 

INTRODUCTION

Metformin, a biguanide, is a well-documented drug used in Type 2 Diabetes Mellitus for over half a century now [1] and is recommended as the drug of choice by the American Diabetes Association [2] and the European Association for the Study of Diabetes [3].

Type 2 Diabetes Mellitus is a chronic disease of adult-onset characterized by high levels of sugar in the blood [4]. Normally, our pancreas produces an important hormone known as insulin, which keeps our blood glucose levels under check [5]. Whenever levels of glucose increase in our blood, for eg after a high carbohydrate meal, insulin synthesis and secretion are accelerated [6] which then acts to increase the uptake of glucose [7] and inhibit biochemical processes involved in the synthesis of glucose [8]. All these actions together help keep our glucose levels in the normal range. However, patients with Type 2 Diabetes Mellitus have a state of ‘insulin resistance’ i.e. their body stops responding to insulin resulting in high blood glucose levels [9]. This excessive glucose gets converted into some toxic products known as advanced glycation end products or AGES [10] which are responsible for the pathogenesis of the various complications associated with diabetes such as cardiovascular disease, nerve damage, kidney disease, eye disease, increased susceptibility to infections etc [11]

Screen Shot 2020-09-03 at 4.26.54 PM.png

Figure 1 showing Insulin Resistance in Patients with Type 2 Diabetes [12]


Metformin has revolutionised the management of Type 2 Diabetes Mellitus by decreasing glucose production from the liver[13], promoting glucose uptake by various cells in the body[14] and delaying glucose absorption from the gut[14]. It also decreases appetite[15] and has a special weight loss promoting effect[16] making it suitable for patients with obesity and Type 2 DM both[17]. 

MOLECULAR ACTION

Metformin is known to be a potent AMPK (adenosine monophosphate‐activated protein kinase)activator[18]. AMPK is an enzyme involved in cellular energy homeostasis[19] and is also a central regulator of fat and glucose metabolism[20]. Upon activation by Metformin, it acts by inhibiting the genes involved in hepatic gluconeogenesis[13] and increases the expression of a glucose transporter named GLUT 4 in fat and muscle cells thereby increasing the uptake of glucose[21]. It was long thought that Metformin acted primarily by the activation of adenosine monophosphate‐activated protein kinase (AMPK) at the molecular level.[22]

 

Screen Shot 2020-09-03 at 4.28.11 PM.png

Figure 2 showing Metformin's downstream actions after AMPK activation[23]

 However, Metformin’s exact mechanism of action has been controversial and the perception that it acts only via AMPK dependent mechanism has changed. A couple of studies revealed that the AMPK pathway is neither essential(Foretz et al) [24] nor sufficient enough [25] to be responsible for the reduced hepatic gluconeogenesis. However, it is debated if the suprapharmolocological doses used in one of these studies[24] had a confounding effect or not as Metformin is known to act through different mechanisms in different concentrations[26]. Even then, increasing data has now accumulated which has suggested or rather proved the existence of some AMPK independent mechanisms[27]. For eg two important mitochondrial actions have been described which along with AMPK mechanism which aid in the suppression of hepatic gluconeogenesis.[28] 

 

Screen Shot 2020-09-03 at 4.29.26 PM.png

Figure 3 illustrating Metformin’s actions to suppress hepatic gluconeogenesis [28]

Interestingly, some studies have strongly indicated that the gut and not the liver is Metformin’s  major site of action unlike what was commonly believed [29 & 30]. This could be supported by the fact that Metformin is known to alter the gut microbiota which leads to increased production of butyrate and propionate, both of which are involved in the maintenance of glucose homeostasis[31]. It also promotes the growth of Akkermansia, an intestinal bacteria, which has been associated with decreased tissue inflammation and improvement in the metabolic profile of diabetics[32]. Moreover, metformin is also known to release incretins such as GLP 1 and also increases the expression of their receptors on the  beta cells of pancreas[33]. Incretin hormones are gut peptides that are secreted after nutrient intake and stimulate insulin secretion together with hyperglycemia.[34]

Screen Shot 2020-09-03 at 4.30.17 PM.png

Figure 4- Metformin’s action on the gut-alters the gut microbiota which leads to increased production of butyrate and propionate both of which are involved in the maintenance of glucose homeostasis, promotes the growth of Akkermansia,induces the release of incretins such as GLP-1,increases expression of GLP-1 receptor on the beta cells of pancreas and retards aging by altering folate and methionine metabolism of intestinal microbiota in C. elegans [35]

Though Metformin’s exact mechanism of action has been a bone of contention, rapid developments are occurring in this regard. Many studies have proposed other novel mechanisms such as inhibition of mitochondrial respiratory-chain complex 1[36], increased FGF21 expression[37], suppression of glucagon signal transduction[38], activation of autophagy[35] , inhibition of  IL‐1β production[39], reduction of terminal endoplasmic reticulum stress[40] to name a few. It is now widely accepted that Metformin brings about its action by acting on various molecular targets and modulating a variety of different pathways and processes. And though we have been using this drug for almost 70 years, new actions continue to be discovered broadening the range of conditions where Metformin could now be used. 

DOSE AND TOXICITY[41] 

Metformin is one of the safest drugs known with a high therapeutic index. Since it has been used for almost half a century now, its safety profile is well documented. FDA recommends a maximum daily dose of 2000mg. Gastric side effects such as nausea and vomiting are common. Lactic acidosis was one of the reasons why there were concerns to approve Metformin initially but now it has been realized that it is a very rare side effect. Contraindications to Metformin include renal failure, hypersensitivity to this drug, and metabolic acidosis.

METFORMIN AND AGING

The world is on the brink of a demographic milestone.The number of people aged 65 or older is projected to grow from an estimated 524 million in 2010 to nearly 1.5 billion in 2050[42].Chronic diseases and illnesses related to this unprecedented aging of humans are a global challenge that poses a risk to handicap world economies and deteriorates the quality of life.[43]Many researchers believe that aging is by far the best predictor of whether people will develop a chronic disease like atherosclerotic heart disease, stroke, cancer, dementia or osteoarthritis.[44]

Screen Shot 2020-09-03 at 4.31.19 PM.png

Figure 5 shows an increase in the incidence of various age-related diseases with increasing age[45]


THE TAME TRIAL

Conventionally, aging has been considered as a debilitating physiological phenomenon that is inevitable.[46] It is believed to be a ‘natural process’ which is bound to happen to all of us. Some researchers believe otherwise and efforts are underway to classify ‘aging’ as a disease and target it as a whole instead of trying to devise treatments for different age-related diseases individually.[47] Targeting this biology of aging, is the famous TAME TRIAL led by the American Federation for Aging Research (AFAR) Scientific Director Nir Barzilai, MD.

The researchers at the TAME trial believe that “The TAME Trial will establish a clinical trial to provide proof-of-concept that aging can be treated, just as we treat diseases. We hope the FDA will approve aging as an indication, to signify that aging can be “treated.” In medical terms, an “indication” for a drug refers to the use of that drug for treating a particular condition.

If aging is made an indication, the TAME Trial will mark a paradigm shift: from treating each age-related medical condition separately, to treating these conditions together, by targeting aging per se.”[47]

The TAME Trial plans to engage around 3,000 adults within the age group of 65-79 all over the US[48]. Though many drugs have shown promising results with respect to aging, Metformin is the main focus because it has been used for over a century as the drug of choice in Type 2 Diabetes Mellitus, has a wide therapeutic window and its safety profile has been very well documented, low cost and, most importantly, various studies have shown the beneficial effects of Metformin in retarding aging and it is also known to modulate key processes that are involved in the pathogenesis of various age-related conditions.[48]

MECHANISM BY WHICH METFORMIN BRINGS ABOUT ITS ANTI AGING EFFECTS

Remarkable progress has taken place in the past few decades in the science and biology of aging. Key pathways implicated in this aging process have been discovered and extensively studied, which have now become a target for intervention.[48] One such pathway is the mTOR pathway (mechanistic target of rapamycin kinase), which has garnered great attention in the anti-aging community. Though the exact mechanism is unknown, mTOR is known to accelerate aging by modulating the hallmarks of aging such as nutrient availability (represented by amino acid availability), energy homeostasis, cellular senescence, cell stemness, and proteostasis.[49]

Screen Shot 2020-09-03 at 4.33.52 PM.png

Figure 6 illustrates the role of the mTOR pathway in the regulation of the hallmarks of aging(black arrows).The depicted hallmarks of aging are also interconnected (grey arrows), suggesting that aging is a coordinated process in which mTOR plays a significant role. [49]

Screen Shot 2020-09-03 at 4.36.10 PM.png

Figure 7 - Growth factors and nutrients activate mTOR which inhibits autophagy and promotes protein synthesis which contributes to cellular stress which might lead to altered cell function, DNA damage,mitochondrial dysfunction etc and induces stem cell exhaustion, which reduces tissue repair and promotes tissue dysfunction which further contribute to the development of age-related diseases. (blue and red) indicates activation,   indicates inhibition.[50]

 Moreover, some studies have suggested Metformin to be a powerful caloric restriction mimetic[51] which acts by activating AMPK [52](known to promote the catabolic reaction and suppresses anabolic reactions).In accordance, studies in C.elegans have shown similar results [53]. Metformin is also known to counteract hyperinsulinemia and reduce IGF-1 levels[54], inhibit mTOR (through both AMPK independent and dependent pathways [55,56], reduce ROS production[57] and even reduce DNA damage[58]. Recent studies have identified some new processes Metformin effects such as the mitohormesis pathway involving PRDX-2 [59]. It also modulates key processes known to have a close association with aging 

and the onset of age-related disorders- inflammation[60], autophagy[61], and even cellular senescence[62]. Metformin has also known to affect two kinases- PKD and MK2, [63] which have an important role to play in cellular stress. 

ANIMAL STUDIES

The unique set of actions that Metformin brings to the table have known to affect aging directly or indirectly. However, most of them have been only reproduced in animal models and not in humans till date and even these animal studies have not shown consistent results. Studies in invertebrate models have shown contrasting results with models such as C.elegans[64] demonstrating positive effects whereas studies in D. [65] have shown quite the opposite. Some review papers have discussed the various confounding factors such as light exposure etc possibly affecting these results[66]. Even the developmental stage at which Metformin was supplemented may also have some effects on the prolongevity.[67]

Figure 8- Summary of the effects of metformin supplementation in invertebrate (Caenorhabditis elegans and Drosophila melanogaster) and vertebrate models (rodents, mainly mice) [68]

CLINICAL STUDIES

Data collected from various animal models have shown promising results. But keep in mind that findings in most animal models may not always translate into humans, more clinical studies are required to come up with definitive results. However, various randomized trials, observational and retrospective studies have shown affirmative results providing evidence for a prolongevity effect which cannot be attributed to glucose-lowering alone. This further substantiates the fact that there are some nonglycemic benefits of Metformin which may hold the key to prolongevity. Various clinical studies have shown improvements in cardiovascular disease [69], stroke[70],cancer[71],autoimmune diseases[72] and neurocognitive disorders[73]. In addition to this, a meta-analysis by Camp et al showed a reduction in all-cause mortality[70]. In fact, a retrospective observational study (Bannister et al) showed that patients with type 2 diabetes initiated with metformin monotherapy had longer survival than their non‐diabetic controls , despite the fact that the diabetic patients were more obese and had greater co-morbidities at baseline.[74]

Screen Shot 2020-09-03 at 4.38.05 PM.png

Figure 9- Metformin and its benefits in various diseases with their proposed mechanism of action[75]

CONCLUSION

Metformin has been used safely for over half a century now and is a drug that the scientific community is well versed with . This makes it the perfect drug to be used and tested for its potential actions in modulating aging and age-related diseases. However, most studies have been done in prediabetics and more clinical trials are required with appropriate controls.[66]

Correlation Is Not Causation, but It’s the Way to Bet

 

References

  1. Inzucchi, S. E., Bergenstal, R. M., Buse, J. B., Diamant, M., Ferrannini, E., Nauck, M., Peters, A. L., Tsapas, A., Wender, R., & Matthews, D. R. (2012). Management of Hyperglycemia in Type 2 Diabetes: A Patient-Centered Approach: Position Statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care, 35(6), 1364–1379. https://doi.org/10.2337/dc12-0413

  2.  Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes—2020. (2019). Diabetes Care, 43(Supplement 1), S98–S110. https://doi.org/10.2337/dc20-s009

  3. Davies, M. J. (2018, December 1). Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. https://care.diabetesjournals.org/lookup/doi/10.2337/dci18-0033

  4. American Diabetes Association (2009). Diagnosis and classification of diabetes mellitus. Diabetes care, 32 Suppl 1(Suppl 1), S62–S67. https://doi.org/10.2337/dc09-S062

  5. Röder, P. V., Wu, B., Liu, Y., & Han, W. (2016). Pancreatic regulation of glucose homeostasis. Experimental & molecular medicine, 48(3), e219. https://doi.org/10.1038/emm.2016.6

  6. Komatsu, M., Takei, M., Ishii, H., & Sato, Y. (2013). Glucose-stimulated insulin secretion: A newer perspective. Journal of diabetes investigation, 4(6), 511–516. https://doi.org/10.1111/jdi.12094

  7. Abel, E. D., Peroni, O., Kim, J. K., Kim, Y. B., Boss, O., Hadro, E., Minnemann, T., Shulman, G. I., & Kahn, B. B. (2001). Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature, 409(6821), 729–733. https://doi.org/10.1038/35055575

  8. Petersen, M. C., & Shulman, G. I. (2018). Mechanisms of Insulin Action and Insulin Resistance. Physiological reviews, 98(4), 2133–2223. https://doi.org/10.1152/physrev.00063.2017

  9. Taylor, R. (2012, April 1). Insulin Resistance and Type 2. Diabetes. https://diabetes.diabetesjournals.org/cgi/doi/10.2337/db12-0073

  10. Chawla, A., Chawla, R., & Jaggi, S. (2016). Microvasular and macrovascular complications in diabetes mellitus: Distinct or continuum?. Indian journal of endocrinology and metabolism, 20(4), 546–551. https://doi.org/10.4103/2230-8210.183480

  11. Deshpande, A. D., Harris-Hayes, M., & Schootman, M. (2008). Epidemiology of diabetes and diabetes-related complications. Physical therapy, 88(11), 1254–1264. https://doi.org/10.2522/ptj.20080020

  12. Type 2 Diabetes. (2019, December). www.healthdirect.gov.au.https://www.healthdirect.gov.au/type-2-diabetes

  13. Triggle, C. R., & Ding, H. (2016). Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiologica, 219(1), 138–151. https://doi.org/10.1111/apha.12644

  14. Collier, C. A., Bruce, C. R., Smith, A. C., Lopaschuk, G., & Dyck, D. J. (2006). Metformin counters the insulin-induced suppression of fatty acid oxidation and stimulation of triacylglycerol storage in rodent skeletal muscle. American Journal of Physiology-Endocrinology and Metabolism, 291(1), E182–E189. https://doi.org/10.1152/ajpendo.00272.2005

  15. Pappachan, J. M., & Viswanath, A. K. (2017). Medical Management of Diabesity: Do We Have Realistic Targets? Current Diabetes Reports, 17(1). https://doi.org/10.1007/s11892-017-0828-9

  16. Lee, A., & Morley, J. E. (1998). Metformin decreases food consumption and induces weight loss in subjects with obesity with type II non-insulin-dependent diabetes. Obesity research, 6(1), 47–53. https://doi.org/10.1002/j.1550-8528.1998.tb00314.x

  17. Kahan, S., & Fujioka, K. (2017). Obesity Pharmacotherapy in Patients With Type 2 Diabetes. Diabetes Spectrum, 30(4), 250–257. https://doi.org/10.2337/ds17-0044

  18. Zhou, G., Myers, R., Li, Y., Chen, Y., Shen, X., Fenyk-Melody, J., Wu, M., Ventre, J., Doebber, T., Fujii, N., Musi, N., Hirshman, M. F., Goodyear, L. J., & Moller, D. E. (2001). Role of AMP-activated protein kinase in mechanism of metformin action. The Journal of clinical investigation, 108(8), 1167–1174. https://doi.org/10.1172/JCI13505

  19. Hardie, D. G., Ross, F. A., & Hawley, S. A. (2012). AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nature reviews. Molecular cell biology, 13(4), 251–262. https://doi.org/10.1038/nrm3311

  20. Towler, M. C., & Hardie, D. G. (2007). AMP-Activated Protein Kinase in Metabolic Control and Insulin Signaling. Circulation Research, 100(3), 328–341. https://doi.org/10.1161/01.res.0000256090.42690.05

  21. Lee, J. O., Lee, S. K., Kim, J. H., Kim, N., You, G. Y., Moon, J. W., Kim, S. J., Park, S. H., & Kim, H. S. (2012). Metformin regulates glucose transporter 4 (GLUT4) translocation through AMP-activated protein kinase (AMPK)-mediated Cbl/CAP signaling in 3T3-L1 preadipocyte cells. The Journal of biological chemistry, 287(53), 44121–44129. https://doi.org/10.1074/jbc.M112.361386

  22. Burcelin, R. (2013). The antidiabetic gutsy role of metformin uncovered? Gut, 63(5), 706–707. https://doi.org/10.1136/gutjnl-2013-305370

  23. Zhou, G., Myers, R., Li, Y., Chen, Y., Shen, X., Fenyk-Melody, J., Wu, M., Ventre, J., Doebber, T., Fujii, N., Musi, N., Hirshman, M. F., Goodyear, L. J., & Moller, D. E. (2001). Role of AMP-activated protein kinase in mechanism of metformin action. The Journal of clinical investigation, 108(8), 1167–1174. https://doi.org/10.1172/JCI13505

  24. Foretz, M., Hébrard, S., Leclerc, J., Zarrinpashneh, E., Soty, M., Mithieux, G., et al. (2010). Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J. Clin. Invest. 120, 2355–2369. doi: 10.1172/JCI40671

  25. Cokorinos, E. C., Delmore, J., Reyes, A. R., Albuquerque, B., Kjøbsted, R., Jørgensen, N. O., et al. (2017). Activation of skeletal muscle AMPK promotes glucose disposal and glucose lowering in non-human primates and mice. Cell Metab. 25, 1147–1159. doi: 10.1016/j.cmet.2017.04.010

  26. He, L., and Wondisford, F. E. (2015). Metformin action: concentrations matter. Cell Metab. 21, 159–162. doi: 10.1016/j.cmet.2015.01.003

  27. Rena, G., Hardie, D. G., & Pearson, E. R. (2017). The mechanisms of action of metformin. Diabetologia, 60(9), 1577–1585. https://doi.org/10.1007/s00125-017-4342-z

  28. An, H., & He, L. (2016). Current understanding of metformin effect on the control of hyperglycemia in diabetes. Journal of Endocrinology, 228(3), R97–R106. https://doi.org/10.1530/joe-15-0447

  29. Natali A, Ferrannini E (2006) Effects of metformin and thiazolidinediones on suppression of hepatic glucose production and stimulation of glucose uptake in type 2 diabetes: a systematic review. Diabetologia 49:434–441

  30. Buse JB, DeFronzo RA, Rosenstock J et al (2016) The primary glucose-lowering effect of metformin resides in the gut, not the circulation: results from short-term pharmacokinetic and 12-week dose-ranging studies. Diabetes Care 39:198–205

  31. Foretz, M., Guigas, B. & Viollet, B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol 15, 569–589 (2019). https://doi.org/10.1038/s41574-019-0242-2

  32. Spruss, A., Kanuri, G., Stahl, C., Bischoff, S. C., & Bergheim, I. (2012). Metformin protects against the development of fructose-induced steatosis in mice: role of the intestinal barrier function. Laboratory investigation; a journal of technical methods and pathology, 92(7), 1020–1032. https://doi.org/10.1038/labinvest.2012.75

  33. Maida, A., Lamont, B. J., Cao, X., & Drucker, D. J. (2011). Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome proliferator-activated receptor-α in mice. Diabetologia, 54(2), 339–349. https://doi.org/10.1007/s00125-010-1937-z

  34. Nauck, M. A., & Meier, J. J. (2018). Incretin hormones: Their role in health and disease. Diabetes, Obesity and Metabolism, 20, 5–21. https://doi.org/10.1111/dom.13129

  35. Hur, K. Y., & Lee, M.-S. (2015). New mechanisms of metformin action: Focusing on mitochondria and the gut. Journal of Diabetes Investigation, 6(6), 600–609. https://doi.org/10.1111/jdi.12328

  36.  El-Mir, M. Y., Nogueira, V., Fontaine, E., Avéret, N., Rigoulet, M., & Leverve, X. (2000). Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. The Journal of biological chemistry, 275(1), 223–228. https://doi.org/10.1074/jbc.275.1.223

  37.  Kim KH, Jeong YT, Kim SH, et al . Metforminâ€induced inhibition of the mitochondrial respiratory chain increases FGF21 expression via ATF4 activation. Biochem Biophys Res Commun 2013; 440: 76–81.

  38.  Miller, R. A., Chu, Q., Xie, J., Foretz, M., Viollet, B., & Birnbaum, M. J. (2013). Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature, 494(7436), 256–260. https://doi.org/10.1038/nature11808

  39. Lee, H. M., Kim, J. J., Kim, H. J., Shong, M., Ku, B. J., & Jo, E. K. (2013). Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes, 62(1), 194–204. https://doi.org/10.2337/db12-0420

  40. Chen, J., Zheng, G., Guo, H., Shi, Z. N., Jiang, J., Wang, X. Y., Yang, X., & Liu, X. Y. (2018). The effect of metformin treatment on endoplasmic reticulum (ER) stress induced by status epilepticus (SE) via the PERK-eIF2α-CHOP pathway. Bosnian journal of basic medical sciences, 18(1), 49–54. https://doi.org/10.17305/bjbms.2017.2044

  41. Crowley MJ, Diamantidis CJ, McDuffie JR, et al. Metformin Use in Patients with Historical Contraindications or Precautions [Internet]. Washington (DC): Department of Veterans Affairs (US); 2016 Sep. APPENDIX A, FDA SAFETY ANNOUNCEMENTS FOR METFORMIN. Available from: https://www.ncbi.nlm.nih.gov/books/NBK409379/

  42. World Population Prospects 2019: Highlights (ST/ESA/SER.A/423). Available at https://population.un.org/wpp/Publications/Files/WPP2019_Highlights.pdf

  43. Lunenfeld, B., & Stratton, P. (2013). The clinical consequences of an ageing world and preventive strategies. Best practice & research. Clinical obstetrics & gynaecology, 27(5), 643–659. https://doi.org/10.1016/j.bpobgyn.2013.02.005

  44. Food, N. (2016, February 2). Pursuing the Dream of Healthy Aging. NDTV Food. https://food.ndtv.com/health/pursuing-the-dream-of-healthy-aging-1272779

  45. Liu, Y., Weng, W., Gao, R., & Liu, Y.(2019). New Insights for Cellular and Molecular Mechanisms of Aging and Aging-Related Diseases: Herbal Medicine as Potential Therapeutic Approach. Oxidative Medicine and Cellular Longevity2019, 1–25. https://doi.org/10.1155/2019/4598167

  46. Franceschi, C., Garagnani, P., Morsiani, C., Conte, M., Santoro, A., Grignolio, A., Monti, D., Capri, M., & Salvioli, S. (2018). The Continuum of Aging and Age-Related Diseases: Common Mechanisms but Different Rates. Frontiers in medicine, 5, 61. https://doi.org/10.3389/fmed.2018.00061

  47. TAME - Targeting Aging with Metformin. (n.d.). American Federation for Aging Research. https://www.afar.org/tame-trial (accessed on July 16  2020)

  48. Barzilai, N., Crandall, J. P., Kritchevsky, S. B., & Espeland, M. A. (2016). Metformin as a Tool to Target Aging. Cell metabolism, 23(6), 1060–1065. https://doi.org/10.1016/j.cmet.2016.05.011

  49. Papadopoli, D., Boulay, K., Kazak, L., Pollak, M., Mallette, F., Topisirovic, I., & Hulea, L. (2019). mTOR as a central regulator of lifespan and aging. F1000Research, 8, F1000 Faculty Rev-998. https://doi.org/10.12688/f1000research.17196.1

  50. Stallone, G., Infante, B., Prisciandaro, C., & Grandaliano, G. (2019). mTOR and Aging: An Old Fashioned Dress. International Journal of Molecular Sciences, 20(11), 2774. doi:10.3390/ijms20112774

  51. Dhahbi JM, Mote PL, Fahy GM, Spindler SR. Identification of potential caloric restriction mimetics by microarray profiling. Physiol Genomics. 2005;23(3):343-350. doi:10.1152/physiolgenomics.00069.2005

  52. Zhou, G.; Goodyear, L.J.; Moller, D.E. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174.

  53. Onken B, Driscoll M. Metformin induces a di- etary restriction-like state and the oxidative stress response to extend C. elegans health- span via AMPK, LKB1, and SKN-1. PLoS One. 2010 Jan;5(1):e8758.

  54. Liu B, Fan Z, Edgerton SM, Yang X, Lind SE, Thor AD. Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle. 2011;10(17):2959–2966.

  55. Howell, J. J., Hellberg, K., Turner, M., Talbott, G., Kolar, M. J., Ross, D. S., Hoxhaj, G., Saghatelian, A., Shaw, R. J., & Manning, B. D. (2017). Metformin Inhibits Hepatic mTORC1 Signaling via Dose-Dependent Mechanisms Involving AMPK and the TSC Complex. Cell metabolism, 25(2), 463–471. https://doi.org/10.1016/j.cmet.2016.12.009

  56. Kalender A, Selvaraj A, Kim SY, Gulati P, Brule S, Viollet B, Kemp BE, Bardeesy N, Dennis P, Schlager JJ, et al. 2010. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab 11: 390–401.

  57. Batandier C, Guigas B, Detaille D, El-Mir MY, Fontaine E, Rigoulet M, Leverve XM. The ROS production induced by a reverse-electron flux at respiratory-chain complex 1 is hampered by metformin. J Bioenerg Bio-membr. 2006; 38:33–42.

  58. Algire C, Moiseeva O, Deschênes-Simard X, Amrein L, Petruccelli L, Birman E, Viollet B, Ferbeyre G, Pollak MN. Metformin reduces endogenous reactive oxygen species and associated DNA damage. Cancer Prev Res (Phila). 2012; 5:536–543. [PubMed: 22262811]

  59. De Haes, W., Frooninckx, L., Van Assche, R., Smolders, A., Depuydt, G., Billen, J., Braeckman, B. P., Schoofs, L., & Temmerman, L. (2014). Metformin promotes lifespan through mitohormesis via the peroxiredoxin PRDX-2. Proceedings of the National Academy of Sciences of the United States of America, 111(24), E2501–E2509. https://doi.org/10.1073/pnas.1321776111

  60. Saisho Y. Metformin and Inflammation: Its Potential Beyond Glucose-lowering Effect. Endocr Metab Immune Disord Drug Targets. 2015; 15:196–205. [PubMed: 25772174]

  61. Song YM, Lee YH, Kim JW, Ham DS, Kang ES, Cha BS, Lee HC, Lee BW. Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy. 2015; 11:46–59.

  62. Moiseeva O, Deschênes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, Bourdeau V, Pollak MN, Ferbeyre G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell. 2013; 12:489–498. [PubMed: 23521863]

  63. Stein, B. D., Calzolari, D., Hellberg, K., Hu, Y. S., He, L., Hung, C.-M., Toyama, E. Q., Ross, D. S., Lillemeier, B. F., Cantley, L. C., Yates, J. R., & Shaw, R. J. (2019). Quantitative In Vivo Proteomics of Metformin Response in Liver Reveals AMPK-Dependent and -Independent Signaling Networks. Cell Reports, 29(10), 3331-3348.e7. https://doi.org/10.1016/j.celrep.2019.10.117

  64. Onken B, Driscoll M. Metformin induces a di- etary restriction-like state and the oxidative stress response to extend C. elegans health- span via AMPK, LKB1, and SKN-1. PLoS One. 2010 Jan;5(1):e8758.

  65. Slack C, Foley A, Partridge L. Activation of AMPK by the putative dietary restriction mimetic metformin is insufficient to extend lifespan in Drosophila. PLoS ONE. 2012; 7:e47699. [PubMed: 23077661]

  66. Glossmann, H. H., & Lutz, O. M. D. (2019). Metformin and Aging: A Review. Gerontology, 65(6), 581–590. https://doi.org/10.1159/000502257

  67. Anisimov VN, Berstein LM, Popovich IG, Zabezhinski MA, Egormin PA, Piskunova TS, Semenchenko AV, Tyndyk ML, Yurova MN, Kovalenko IG, et al. 2011. If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging 3: 148–157.

  68.  Novelle, M. G., Ali, A., Diéguez, C., Bernier, M., & de Cabo, R. (2016). Metformin: A Hopeful Promise in Aging Research. Cold Spring Harbor perspectives in medicine, 6(3), a025932. https://doi.org/10.1101/cshperspect.a025932

  69. Diabetes Prevention Program (DPP) Research Group (2002). The Diabetes Prevention Program (DPP): description of lifestyle intervention. Diabetes care, 25(12), 2165–2171. https://doi.org/10.2337/diacare.25.12.2165

  70.  Campbell, J. M., Bellman, S. M., Stephenson, M. D., & Lisy., K. (2017). Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: A systematic review and meta-analysis. Ageing Research Reviews, 40, 31–44. https://doi.org/10.1016/j.arr.2017.08.003

  71. Gandini, S., Puntoni, M., Heckman-Stoddard, B. M., Dunn, B. K., Ford, L., DeCensi, A., & Szabo, E. (2014). Metformin and Cancer Risk and Mortality: A Systematic Review and Meta-analysis Taking into Account Biases and Confounders. Cancer Prevention Research, 7(9), 867–885. https://doi.org/10.1158/1940-6207.capr-13-0424

  72.  Ursini, F., Russo, E., Pellino, G., D’Angelo, S., Chiaravalloti, A., De Sarro, G., Manfredini, R., & De Giorgio, R. (2018). Metformin and Autoimmunity: A “New Deal” of an Old Drug. Frontiers in Immunology, 9, 231–300. https://doi.org/10.3389/fimmu.2018.01236

  73.  Ng, T. P., Feng, L., Yap, K. B., Lee, T. S., Tan, C. H., & Winblad, B. (2014). Long-term metformin usage and cognitive function among older adults with diabetes. Journal of Alzheimer's disease : JAD, 41(1), 61–68. https://doi.org/10.3233/JAD-131901

  74. Bannister, C. A., Holden, S. E., Jenkins-Jones, S., Morgan, C. L., Halcox, J. P., Schernthaner, G., Mukherjee, J., & Currie, C. J. (2014). Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls. Diabetes, Obesity and Metabolism, 16(11), 1165–1173. https://doi.org/10.1111/dom.12354

  75. Lv, Z., & Guo, Y. (2020). Metformin and Its Benefits for Various Diseases. Frontiers in Endocrinology, 11, https://doi.org/10.3389/fendo.2020.00191

Previous
Previous

Next
Next