Diabetes Metab J.  2024 Jan;48(1):1-18. 10.4093/dmj.2023.0115.

Mitochondrial Stress and Mitokines: Therapeutic Perspectives for the Treatment of Metabolic Diseases

Affiliations
  • 1Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
  • 2Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
  • 3Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea

Abstract

Mitochondrial stress and the dysregulated mitochondrial unfolded protein response (UPRmt) are linked to various diseases, including metabolic disorders, neurodegenerative diseases, and cancer. Mitokines, signaling molecules released by mitochondrial stress response and UPRmt, are crucial mediators of inter-organ communication and influence systemic metabolic and physiological processes. In this review, we provide a comprehensive overview of mitokines, including their regulation by exercise and lifestyle interventions and their implications for various diseases. The endocrine actions of mitokines related to mitochondrial stress and adaptations are highlighted, specifically the broad functions of fibroblast growth factor 21 and growth differentiation factor 15, as well as their specific actions in regulating inter-tissue communication and metabolic homeostasis. Finally, we discuss the potential of physiological and genetic interventions to reduce the hazards associated with dysregulated mitokine signaling and preserve an equilibrium in mitochondrial stress-induced responses. This review provides valuable insights into the mechanisms underlying mitochondrial regulation of health and disease by exploring mitokine interactions and their regulation, which will facilitate the development of targeted therapies and personalized interventions to improve health outcomes and quality of life.

Keyword

Fibroblast growth factor 21; Growth differentiation factor 15; Metabolic diseases; Mitochondria; Unfolded protein response

Figure

  • Fig. 1. Regulation of fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) expression by mitochondrial stress response (MSR) and mitochondrial unfolded protein response (UPRmt). The integrated stress response (ISR) is a cellular pathway activated in response to mitochondrial defects. In this pathway, a molecule called DAP3 binding cell death enhancer (DELE) plays a crucial role. DELE activates the heme-regulated inhibitor (HRI), which leads to the phosphorylation of eukaryotic initiation factor 2α (p-eIF2α) or triggers the ISR directly. This activation of the ISR facilitates the expression of key regulatory factors, such as activating transcription factors 4 (ATF4), ATF5, and C/EBP homologous protein (CHOP), which act as crucial regulators for the expression of FGF21 and GDF15. Additionally, an elevated ratio of adenosine monophosphate (AMP)/adenosine triphosphate (ATP), signaling energy stress, can induce the activation of AMP-activated protein kinase (AMPK). This activation occurs through a potential AKT serine/threonine kinase 1 (AKT1)-mediated mechanism. Ultimately, the activation of AMPK stimulates the expression of FGF21 or GDF15, further contributing to the regulatory processes involved in MSR and UPRmt. ETC, electron transport chain.

  • Fig. 2. The pathophysiological roles of fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) in the metabolic disease. FGF21 regulates appetite, enhances ketogenesis, improves insulin sensitivity, promotes lipolysis, and influences glucose homeostasis. GDF15 regulates appetite, leads to weight loss, decreases muscle mass, promotes lipolysis and fatty acid oxidation, and affects liver metabolism.


Reference

1. McBride HM, Neuspiel M, Wasiak S. Mitochondria: more than just a powerhouse. Curr Biol. 2006; 16:R551–60.
2. Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell. 2012; 148:1145–59.
3. Williams AS, Koves TR, Davidson MT, Crown SB, Fisher-Wellman KH, Torres MJ, et al. Disruption of acetyl-lysine turnover in muscle mitochondria promotes insulin resistance and redox stress without overt respiratory dysfunction. Cell Metab. 2020; 31:131–47.
4. Picard M, Shirihai OS. Mitochondrial signal transduction. Cell Metab. 2022; 34:1620–53.
5. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022; 375:1254–61.
6. Shadel GS, Horvath TL. Mitochondrial ROS signaling in organismal homeostasis. Cell. 2015; 163:560–9.
7. Wredenberg A, Wibom R, Wilhelmsson H, Graff C, Wiener HH, Burden SJ, et al. Increased mitochondrial mass in mitochondrial myopathy mice. Proc Natl Acad Sci U S A. 2002; 99:15066–71.
8. Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener. 2020; 15:30.
9. Hammerschmidt P, Ostkotte D, Nolte H, Gerl MJ, Jais A, Brunner HL, et al. CerS6-derived sphingolipids interact with Mff and promote mitochondrial fragmentation in obesity. Cell. 2019; 177:1536–52.
10. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006; 443:787–95.
11. Zong WX, Rabinowitz JD, White E. Mitochondria and cancer. Mol Cell. 2016; 61:667–76.
12. Booth DM, Varnai P, Joseph SK, Hajnoczky G. Oxidative bursts of single mitochondria mediate retrograde signaling toward the ER. Mol Cell. 2021; 81:3866–76.
13. Bar-Ziv R, Bolas T, Dillin A. Systemic effects of mitochondrial stress. EMBO Rep. 2020; 21:e50094.
14. Nicolas-Avila JA, Lechuga-Vieco AV, Esteban-Martinez L, Sanchez-Diaz M, Diaz-Garcia E, Santiago DJ, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. 2020; 183:94–109.
15. Gan Z, Fu T, Kelly DP, Vega RB. Skeletal muscle mitochondrial remodeling in exercise and diseases. Cell Res. 2018; 28:969–80.
16. Chavez JA, Summers SA. A ceramide-centric view of insulin resistance. Cell Metab. 2012; 15:585–94.
17. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002; 51:2944–50.
18. Pedersen BK. Physical activity and muscle-brain crosstalk. Nat Rev Endocrinol. 2019; 15:383–92.
19. Aguiar AS Jr, Tuon T, Soares FS, da Rocha LG, Silveira PC, Pinho RA. The effect of n-acetylcysteine and deferoxamine on exercise-induced oxidative damage in striatum and hippocampus of mice. Neurochem Res. 2008; 33:729–36.
20. Nargund AM, Pellegrino MW, Fiorese CJ, Baker BM, Haynes CM. Mitochondrial import efficiency of ATFS-1 regulates mitochondrial UPR activation. Science. 2012; 337:587–90.
21. Mohrin M, Shin J, Liu Y, Brown K, Luo H, Xi Y, et al. Stem cell aging: a mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science. 2015; 347:1374–7.
22. Tyynismaa H, Mjosund KP, Wanrooij S, Lappalainen I, Ylikallio E, Jalanko A, et al. Mutant mitochondrial helicase twinkle causes multiple mtDNA deletions and a late-onset mitochondrial disease in mice. Proc Natl Acad Sci U S A. 2005; 102:17687–92.
23. Lee YG, Park DH, Chae YC. Role of mitochondrial stress response in cancer progression. Cells. 2022; 11:771.
24. Deng P, Haynes CM. Mitochondrial dysfunction in cancer: potential roles of ATF5 and the mitochondrial UPR. Semin Cancer Biol. 2017; 47:43–9.
25. Haynes CM, Ron D. The mitochondrial UPR: protecting organelle protein homeostasis. J Cell Sci. 2010; 123(Pt 22):3849–55.
26. Ruan L, Zhou C, Jin E, Kucharavy A, Zhang Y, Wen Z, et al. Cytosolic proteostasis through importing of misfolded proteins into mitochondria. Nature. 2017; 543:443–6.
27. Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. 2020; 21:421–38.
28. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004; 18:3066–77.
29. Conte M, Martucci M, Chiariello A, Franceschi C, Salvioli S. Mitochondria, immunosenescence and inflammaging: a role for mitokines? Semin Immunopathol. 2020; 42:607–17.
30. Inigo JR, Chandra D. The mitochondrial unfolded protein response (UPRmt): shielding against toxicity to mitochondria in cancer. J Hematol Oncol. 2022; 15:98.
31. Zhao Q, Wang J, Levichkin IV, Stasinopoulos S, Ryan MT, Hoogenraad NJ. A mitochondrial specific stress response in mammalian cells. EMBO J. 2002; 21:4411–9.
32. Papa L, Germain D. Estrogen receptor mediates a distinct mitochondrial unfolded protein response. J Cell Sci. 2011; 124(Pt 9):1396–402.
33. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Canto C, et al. The NAD(+)/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. 2013; 154:430–41.
34. Houtkooper RH, Mouchiroud L, Ryu D, Moullan N, Katsyuba E, Knott G, et al. Mitonuclear protein imbalance as a conserved longevity mechanism. Nature. 2013; 497:451–7.
35. Chen PX, Zhang L, Chen D, Tian Y. Mitochondrial stress and aging: lessons from C. elegans. Semin Cell Dev Biol. 2024; 154(Pt A):69–76.
36. Lionaki E, Gkikas I, Daskalaki I, Ioannidi MK, Klapa MI, Tavernarakis N. Mitochondrial protein import determines lifespan through metabolic reprogramming and de novo serine biosynthesis. Nat Commun. 2022; 13:651.
37. Tian Y, Garcia G, Bian Q, Steffen KK, Joe L, Wolff S, et al. Mitochondrial stress induces chromatin reorganization to promote longevity and UPR(mt). Cell. 2016; 165:1197–208.
38. Rainbolt TK, Atanassova N, Genereux JC, Wiseman RL. Stressregulated translational attenuation adapts mitochondrial protein import through Tim17A degradation. Cell Metab. 2013; 18:908–19.
39. Pellegrino MW, Nargund AM, Kirienko NV, Gillis R, Fiorese CJ, Haynes CM. Mitochondrial UPR-regulated innate immunity provides resistance to pathogen infection. Nature. 2014; 516:414–7.
40. Kim HE, Grant AR, Simic MS, Kohnz RA, Nomura DK, Durieux J, et al. Lipid biosynthesis coordinates a mitochondrialto-cytosolic stress response. Cell. 2016; 166:1539–52.
41. Livingston MJ, Wang J, Zhou J, Wu G, Ganley IG, Hill JA, et al. Clearance of damaged mitochondria via mitophagy is important to the protective effect of ischemic preconditioning in kidneys. Autophagy. 2019; 15:2142–62.
42. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007; 8:519–29.
43. Shpilka T, Haynes CM. The mitochondrial UPR: mechanisms, physiological functions and implications in ageing. Nat Rev Mol Cell Biol. 2018; 19:109–20.
44. Klaus S, Ost M. Mitochondrial uncoupling and longevity: a role for mitokines? Exp Gerontol. 2020; 130:110796.
45. Cox CS, McKay SE, Holmbeck MA, Christian BE, Scortea AC, Tsay AJ, et al. Mitohormesis in mice via sustained basal activation of mitochondrial and antioxidant signaling. Cell Metab. 2018; 28:776–86.
46. Palikaras K, Lionaki E, Tavernarakis N. Coordination of mitophagy and mitochondrial biogenesis during ageing in C. elegans. Nature. 2015; 521:525–8.
47. Song T, Song X, Zhu C, Patrick R, Skurla M, Santangelo I, et al. Mitochondrial dysfunction, oxidative stress, neuroinflammation, and metabolic alterations in the progression of Alzheimer’s disease: a meta-analysis of in vivo magnetic resonance spectroscopy studies. Ageing Res Rev. 2021; 72:101503.
48. Wang SF, Chen S, Tseng LM, Lee HC. Role of the mitochondrial stress response in human cancer progression. Exp Biol Med (Maywood). 2020; 245:861–78.
49. Patti ME, Corvera S. The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr Rev. 2010; 31:364–95.
50. Politis-Barber V, Brunetta HS, Paglialunga S, Petrick HL, Holloway GP. Long-term, high-fat feeding exacerbates short-term increases in adipose mitochondrial reactive oxygen species, without impairing mitochondrial respiration. Am J Physiol Endocrinol Metab. 2020; 319:E376–87.
51. Mansouri A, Gattolliat CH, Asselah T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology. 2018; 155:629–47.
52. Bhatia D, Capili A, Choi ME. Mitochondrial dysfunction in kidney injury, inflammation, and disease: potential therapeutic approaches. Kidney Res Clin Pract. 2020; 39:244–58.
53. Li X, Zhang W, Cao Q, Wang Z, Zhao M, Xu L, et al. Mitochondrial dysfunction in fibrotic diseases. Cell Death Discov. 2020; 6:80.
54. Chung HK, Kim JT, Kim HW, Kwon M, Kim SY, Shong M, et al. GDF15 deficiency exacerbates chronic alcohol- and carbon tetrachloride-induced liver injury. Sci Rep. 2017; 7:17238.
55. Lu B, Yadav S, Shah PG, Liu T, Tian B, Pukszta S, et al. Roles for the human ATP-dependent Lon protease in mitochondrial DNA maintenance. J Biol Chem. 2007; 282:17363–74.
56. Gibellini L, De Gaetano A, Mandrioli M, Van Tongeren E, Bortolotti CA, Cossarizza A, et al. The biology of Lonp1: more than a mitochondrial protease. Int Rev Cell Mol Biol. 2020; 354:1–61.
57. Guo X, Aviles G, Liu Y, Tian R, Unger BA, Lin YT, et al. Mitochondrial stress is relayed to the cytosol by an OMA1-DELE1-HRI pathway. Nature. 2020; 579:427–32.
58. Lee H, Ha TY, Jung CH, Nirmala FS, Park SY, Huh YH, et al. Mitochondrial dysfunction in skeletal muscle contributes to the development of acute insulin resistance in mice. J Cachexia Sarcopenia Muscle. 2021; 12:1925–39.
59. Yan J, Jiang J, He L, Chen L. Mitochondrial superoxide/hydrogen peroxide: an emerging therapeutic target for metabolic diseases. Free Radic Biol Med. 2020; 152:33–42.
60. Martinez BA, Petersen DA, Gaeta AL, Stanley SP, Caldwell GA, Caldwell KA. Dysregulation of the mitochondrial unfolded protein response induces non-apoptotic dopaminergic neurodegeneration in C. elegans models of Parkinson’s disease. J Neurosci. 2017; 37:11085–100.
61. Kumar R, Chaudhary AK, Woytash J, Inigo JR, Gokhale AA, Bshara W, et al. A mitochondrial unfolded protein response inhibitor suppresses prostate cancer growth in mice via HSP60. J Clin Invest. 2022; 132:e149906.
62. Burtscher J, Soltany A, Visavadiya NP, Burtscher M, Millet GP, Khoramipour K, et al. Mitochondrial stress and mitokines in aging. Aging Cell. 2023; 22:e13770.
63. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med. 2013; 19:83–92.
64. Beydoun MA, Noren Hooten N, Weiss J, Beydoun HA, Georgescu M, Freeman DW, et al. GDF15 and its association with cognitive performance over time in a longitudinal study of middle-aged urban adults. Brain Behav Immun. 2023; 108:340–9.
65. Durieux J, Wolff S, Dillin A. The cell-non-autonomous nature of electron transport chain-mediated longevity. Cell. 2011; 144:79–91.
66. Whitham M, Febbraio MA. The ever-expanding myokinome: discovery challenges and therapeutic implications. Nat Rev Drug Discov. 2016; 15:719–29.
67. Ost M, Coleman V, Voigt A, van Schothorst EM, Keipert S, van der Stelt I, et al. Muscle mitochondrial stress adaptation operates independently of endogenous FGF21 action. Mol Metab. 2015; 5:79–90.
68. Zhang IW, Curto A, Lopez-Vicario C, Casulleras M, Duran-Guell M, Flores-Costa R, et al. Mitochondrial dysfunction governs immunometabolism in leukocytes of patients with acute-on-chronic liver failure. J Hepatol. 2022; 76:93–106.
69. Conte M, Martucci M, Mosconi G, Chiariello A, Cappuccilli M, Totti V, et al. GDF15 plasma level is inversely associated with level of physical activity and correlates with markers of inflammation and muscle weakness. Front Immunol. 2020; 11:915.
70. Lertpatipanpong P, Lee J, Kim I, Eling T, Oh SY, Seong JK, et al. The anti-diabetic effects of NAG-1/GDF15 on HFD/STZ-induced mice. Sci Rep. 2021; 11:15027.
71. Klein AB, Nicolaisen TS, Ortenblad N, Gejl KD, Jensen R, Fritzen AM, et al. Pharmacological but not physiological GDF15 suppresses feeding and the motivation to exercise. Nat Commun. 2021; 12:1041.
72. Xiong Y, Walker K, Min X, Hale C, Tran T, Komorowski R, et al. Long-acting MIC-1/GDF15 molecules to treat obesity: evidence from mice to monkeys. Sci Transl Med. 2017; 9:eaan8732.
73. Lee C, Zeng J, Drew BG, Sallam T, Martin-Montalvo A, Wan J, et al. The mitochondrial-derived peptide MOTS-c promotes metabolic homeostasis and reduces obesity and insulin resistance. Cell Metab. 2015; 21:443–54.
74. Watanabe N, Nakano M, Mitsuishi Y, Hara N, Mano T, Iwata A, et al. Transcriptional downregulation of FAM3C/ILEI in the Alzheimer’s brain. Hum Mol Genet. 2021; 31:122–32.
75. Kang SG, Choi MJ, Jung SB, Chung HK, Chang JY, Kim JT, et al. Differential roles of GDF15 and FGF21 in systemic metabolic adaptation to the mitochondrial integrated stress response. iScience. 2021; 24:102181.
76. Oost LJ, Kustermann M, Armani A, Blaauw B, Romanello V. Fibroblast growth factor 21 controls mitophagy and muscle mass. J Cachexia Sarcopenia Muscle. 2019; 10:630–42.
77. Patel MS, Lee J, Baz M, Wells CE, Bloch S, Lewis A, et al. Growth differentiation factor-15 is associated with muscle mass in chronic obstructive pulmonary disease and promotes muscle wasting in vivo. J Cachexia Sarcopenia Muscle. 2016; 7:436–48.
78. Berendzen KM, Durieux J, Shao LW, Tian Y, Kim HE, Wolff S, et al. Neuroendocrine coordination of mitochondrial stress signaling and proteostasis. Cell. 2016; 166:1553–63.
79. Shao LW, Niu R, Liu Y. Neuropeptide signals cell non-autonomous mitochondrial unfolded protein response. Cell Res. 2016; 26:1182–96.
80. Zhang Q, Wu X, Chen P, Liu L, Xin N, Tian Y, et al. The mitochondrial unfolded protein response is mediated cell-non-autonomously by retromer-dependent Wnt signaling. Cell. 2018; 174:870–83.
81. Kim SJ, Guerrero N, Wassef G, Xiao J, Mehta HH, Cohen P, et al. The mitochondrial-derived peptide humanin activates the ERK1/2, AKT, and STAT3 signaling pathways and has age-dependent signaling differences in the hippocampus. Oncotarget. 2016; 7:46899–912.
82. Lee C, Kim KH, Cohen P. MOTS-c: a novel mitochondrialderived peptide regulating muscle and fat metabolism. Free Radic Biol Med. 2016; 100:182–7.
83. Kang SG, Yi HS, Choi MJ, Ryu MJ, Jung S, Chung HK, et al. ANGPTL6 expression is coupled with mitochondrial OXPHOS function to regulate adipose FGF21. J Endocrinol. 2017; 233:105–18.
84. Rose G, Santoro A, Salvioli S. Mitochondria and mitochondria-induced signalling molecules as longevity determinants. Mech Ageing Dev. 2017; 165(Pt B):115–28.
85. Suomalainen A, Elo JM, Pietilainen KH, Hakonen AH, Sevastianova K, Korpela M, et al. FGF-21 as a biomarker for muscle-manifesting mitochondrial respiratory chain deficiencies: a diagnostic study. Lancet Neurol. 2011; 10:806–18.
86. Pereira RO, Tadinada SM, Zasadny FM, Oliveira KJ, Pires KM, Olvera A, et al. OPA1 deficiency promotes secretion of FGF21 from muscle that prevents obesity and insulin resistance. EMBO J. 2017; 36:2126–45.
87. Tezze C, Romanello V, Desbats MA, Fadini GP, Albiero M, Favaro G, et al. Age-associated loss of OPA1 in muscle impacts muscle mass, metabolic homeostasis, systemic inflammation, and epithelial senescence. Cell Metab. 2017; 25:1374–89.
88. Keipert S, Ost M, Johann K, Imber F, Jastroch M, van Schothorst EM, et al. Skeletal muscle mitochondrial uncoupling drives endocrine cross-talk through the induction of FGF21 as a myokine. Am J Physiol Endocrinol Metab. 2014; 306:E469–82.
89. Fiorese CJ, Schulz AM, Lin YF, Rosin N, Pellegrino MW, Haynes CM. The transcription factor ATF5 mediates a mammalian mitochondrial UPR. Curr Biol. 2016; 26:2037–43.
90. Khan NA, Nikkanen J, Yatsuga S, Jackson C, Wang L, Pradhan S, et al. mTORC1 regulates mitochondrial integrated stress response and mitochondrial myopathy progression. Cell Metab. 2017; 26:419–28.
91. Salminen A, Kauppinen A, Kaarniranta K. FGF21 activates AMPK signaling: impact on metabolic regulation and the aging process. J Mol Med (Berl). 2017; 95:123–31.
92. Tyynismaa H, Carroll CJ, Raimundo N, Ahola-Erkkila S, Wenz T, Ruhanen H, et al. Mitochondrial myopathy induces a starvation-like response. Hum Mol Genet. 2010; 19:3948–58.
93. Hondares E, Rosell M, Diaz-Delfin J, Olmos Y, Monsalve M, Iglesias R, et al. Peroxisome proliferator-activated receptor α (PPARα) induces PPARγ coactivator 1α (PGC-1α) gene expression and contributes to thermogenic activation of brown fat: involvement of PRDM16. J Biol Chem. 2011; 286:43112–22.
94. Li X, Hong Y, He H, Jiang G, You W, Liang X, et al. FGF21 mediates mesenchymal stem cell senescence via regulation of mitochondrial dynamics. Oxid Med Cell Longev. 2019; 2019:4915149.
95. Chau MD, Gao J, Yang Q, Wu Z, Gromada J. Fibroblast growth factor 21 regulates energy metabolism by activating the AMPKSIRT1-PGC-1alpha pathway. Proc Natl Acad Sci U S A. 2010; 107:12553–8.
96. Kharitonenkov A, DiMarchi R. FGF21 revolutions: recent advances illuminating FGF21 biology and medicinal properties. Trends Endocrinol Metab. 2015; 26:608–17.
97. Owen BM, Mangelsdorf DJ, Kliewer SA. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends Endocrinol Metab. 2015; 26:22–9.
98. Ruhlmann C, Dannehl D, Brodtruck M, Adams AC, Stenzel J, Lindner T, et al. Neuroprotective effects of the FGF21 analogue LY2405319. J Alzheimers Dis. 2021; 80:357–69.
99. Fang X, Ma J, Mu D, Li B, Lian B, Sun C. FGF21 protects dopaminergic neurons in Parkinson’s disease models via repression of neuroinflammation. Neurotox Res. 2020; 37:616–27.
100. Liang Q, Zhong L, Zhang J, Wang Y, Bornstein SR, Triggle CR, et al. FGF21 maintains glucose homeostasis by mediating the cross talk between liver and brain during prolonged fasting. Diabetes. 2014; 63:4064–75.
101. Battaglia CR, Cursano S, Calzia E, Catanese A, Boeckers TM. Corticotropin-releasing hormone (CRH) alters mitochondrial morphology and function by activating the NF-kB-DRP1 axis in hippocampal neurons. Cell Death Dis. 2020; 11:1004.
102. Maratos-Flier E. Fatty liver and FGF21 physiology. Exp Cell Res. 2017; 360:2–5.
103. Potthoff MJ. FGF21 and metabolic disease in 2016: a new frontier in FGF21 biology. Nat Rev Endocrinol. 2017; 13:74–6.
104. Dunshee DR, Bainbridge TW, Kljavin NM, Zavala-Solorio J, Schroeder AC, Chan R, et al. Fibroblast activation protein cleaves and inactivates fibroblast growth factor 21. J Biol Chem. 2016; 291:5986–96.
105. Zhen EY, Jin Z, Ackermann BL, Thomas MK, Gutierrez JA. Circulating FGF21 proteolytic processing mediated by fibroblast activation protein. Biochem J. 2016; 473:605–14.
106. Sanyal A, Charles ED, Neuschwander-Tetri BA, Loomba R, Harrison SA, Abdelmalek MF, et al. Pegbelfermin (BMS986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet. 2019; 392:2705–17.
107. Harrison SA, Ruane PJ, Freilich BL, Neff G, Patil R, Behling CA, et al. Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nat Med. 2021; 27:1262–71.
108. Talukdar S, Zhou Y, Li D, Rossulek M, Dong J, Somayaji V, et al. A long-acting FGF21 molecule, PF-05231023, decreases body weight and improves lipid profile in non-human primates and type 2 diabetic subjects. Cell Metab. 2016; 23:427–40.
109. Charles ED, Neuschwander-Tetri BA, Pablo Frias J, Kundu S, Luo Y, Tirucherai GS, et al. Pegbelfermin (BMS-986036), PEGylated FGF21, in patients with obesity and type 2 diabetes: results from a randomized phase 2 study. Obesity (Silver Spring). 2019; 27:41–9.
110. Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, et al. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab. 2013; 18:333–40.
111. Kim AM, Somayaji VR, Dong JQ, Rolph TP, Weng Y, Chabot JR, et al. Once-weekly administration of a long-acting fibroblast growth factor 21 analogue modulates lipids, bone turnover markers, blood pressure and body weight differently in obese people with hypertriglyceridaemia and in non-human primates. Diabetes Obes Metab. 2017; 19:1762–72.
112. Kaufman A, Abuqayyas L, Denney WS, Tillman EJ, Rolph T. AKR-001, an Fc-FGF21 analog, showed sustained pharmacodynamic effects on insulin sensitivity and lipid metabolism in type 2 diabetes patients. Cell Rep Med. 2020; 1:100057.
113. Brown EA, Minnich A, Sanyal AJ, Loomba R, Du S, Schwarz J, et al. Effect of pegbelfermin on NASH and fibrosis-related biomarkers and correlation with histological response in the FALCON 1 trial. JHEP Rep. 2023; 5:100661.
114. Kim KH, Jeong YT, Kim SH, Jung HS, Park KS, Lee HY, et al. Metformin-induced inhibition of the mitochondrial respiratory chain increases FGF21 expression via ATF4 activation. Biochem Biophys Res Commun. 2013; 440:76–81.
115. Muise ES, Azzolina B, Kuo DW, El-Sherbeini M, Tan Y, Yuan X, et al. Adipose fibroblast growth factor 21 is up-regulated by peroxisome proliferator-activated receptor gamma and altered metabolic states. Mol Pharmacol. 2008; 74:403–12.
116. Yatsuga S, Fujita Y, Ishii A, Fukumoto Y, Arahata H, Kakuma T, et al. Growth differentiation factor 15 as a useful biomarker for mitochondrial disorders. Ann Neurol. 2015; 78:814–23.
117. Montero R, Yubero D, Villarroya J, Henares D, Jou C, Rodriguez MA, et al. GDF-15 is elevated in children with mitochondrial diseases and is induced by mitochondrial dysfunction. PLoS One. 2016; 11:e0148709.
118. Fujita Y, Ito M, Kojima T, Yatsuga S, Koga Y, Tanaka M. GDF15 is a novel biomarker to evaluate efficacy of pyruvate therapy for mitochondrial diseases. Mitochondrion. 2015; 20:34–42.
119. Koene S, de Laat P, van Tienoven DH, Weijers G, Vriens D, Sweep FC, et al. Serum GDF15 levels correlate to mitochondrial disease severity and myocardial strain, but not to disease progression in adult m.3243A>G carriers. JIMD Rep. 2015; 24:69–81.
120. Koopman WJ, Beyrath J, Fung CW, Koene S, Rodenburg RJ, Willems PH, et al. Mitochondrial disorders in children: toward development of small-molecule treatment strategies. EMBO Mol Med. 2016; 8:311–27.
121. Mottis A, Herzig S, Auwerx J. Mitocellular communication: shaping health and disease. Science. 2019; 366:827–32.
122. Tezze C, Romanello V, Sandri M. FGF21 as modulator of metabolism in health and disease. Front Physiol. 2019; 10:419.
123. Johann K, Kleinert M, Klaus S. The role of GDF15 as a myomitokine. Cells. 2021; 10:2990.
124. Tsai VW, Husaini Y, Sainsbury A, Brown DA, Breit SN. The MIC-1/GDF15-GFRAL pathway in energy homeostasis: implications for obesity, cachexia, and other associated diseases. Cell Metab. 2018; 28:353–68.
125. Suarez-Rivero JM, Pastor-Maldonado CJ, Povea-Cabello S, Alvarez-Cordoba M, Villalon-Garcia I, Talaveron-Rey M, et al. Activation of the mitochondrial unfolded protein response: a new therapeutic target? Biomedicines. 2022; 10:1611.
126. Costa-Mattioli M, Walter P. The integrated stress response: from mechanism to disease. Science. 2020; 368:eaat5314.
127. Patel S, Alvarez-Guaita A, Melvin A, Rimmington D, Dattilo A, Miedzybrodzka EL, et al. GDF15 provides an endocrine signal of nutritional stress in mice and humans. Cell Metab. 2019; 29:707–18.
128. Miyake M, Zhang J, Yasue A, Hisanaga S, Tsugawa K, Sakaue H, et al. Integrated stress response regulates GDF15 secretion from adipocytes, preferentially suppresses appetite for a high-fat diet and improves obesity. iScience. 2021; 24:103448.
129. Yang L, Chang CC, Sun Z, Madsen D, Zhu H, Padkaer SB, et al. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nat Med. 2017; 23:1158–66.
130. Mullican SE, Lin-Schmidt X, Chin CN, Chavez JA, Furman JL, Armstrong AA, et al. GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Nat Med. 2017; 23:1150–7.
131. Emmerson PJ, Wang F, Du Y, Liu Q, Pickard RT, Gonciarz MD, et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat Med. 2017; 23:1215–9.
132. Hsu JY, Crawley S, Chen M, Ayupova DA, Lindhout DA, Higbee J, et al. Non-homeostatic body weight regulation through a brainstem-restricted receptor for GDF15. Nature. 2017; 550:255–9.
133. Chow CF, Guo X, Asthana P, Zhang S, Wong SK, Fallah S, et al. Body weight regulation via MT1-MMP-mediated cleavage of GFRAL. Nat Metab. 2022; 4:203–12.
134. Fung E, Kang L, Sapashnik D, Benard S, Sievers A, Liu Y, et al. Fc-GDF15 glyco-engineering and receptor binding affinity optimization for body weight regulation. Sci Rep. 2021; 11:8921.
135. Zhang Y, Zhao X, Dong X, Zhang Y, Zou H, Jin Y, et al. Activity-balanced GLP-1/GDF15 dual agonist reduces body weight and metabolic disorder in mice and non-human primates. Cell Metab. 2023; 35:287–98.
136. Benichou O, Coskun T, Gonciarz MD, Garhyan P, Adams AC, Du Y, et al. Discovery, development, and clinical proof of mechanism of LY3463251, a long-acting GDF15 receptor agonist. Cell Metab. 2023; 35:274–86.
137. Fejzo MS, Arzy D, Tian R, MacGibbon KW, Mullin PM. Evidence GDF15 plays a role in familial and recurrent hyperemesis gravidarum. Geburtshilfe Frauenheilkd. 2018; 78:866–70.
138. Breen DM, Kim H, Bennett D, Calle RA, Collins S, Esquejo RM, et al. GDF-15 neutralization alleviates platinum-based chemotherapy-induced emesis, anorexia, and weight loss in mice and nonhuman primates. Cell Metab. 2020; 32:938–50.
139. Kim-Muller JY, Song L, LaCarubba Paulhus B, Pashos E, Li X, Rinaldi A, et al. GDF15 neutralization restores muscle function and physical performance in a mouse model of cancer cachexia. Cell Rep. 2023; 42:111947.
140. Jain R, Kim EJ, Lenz HJ, Messersmith WA, Picozzi VJ, Beg MS, et al. 550P Initial results of a phase Ia/Ib study of NGM120, a first-in-class anti-GDNF family receptor alpha like (GFRAL) antibody in patients (pts) with advanced solid tumors. Ann Oncol. 2021; 32(S5):S610–1.
141. Kong BS, Lee C, Cho YM. Mitochondrial-encoded peptide MOTS-c, diabetes, and aging-related diseases. Diabetes Metab J. 2023; 47:315–24.
142. Saghatelian A, Couso JP. Discovery and characterization of smORF-encoded bioactive polypeptides. Nat Chem Biol. 2015; 11:909–16.
143. Miller B, Kim SJ, Kumagai H, Yen K, Cohen P. Mitochondriaderived peptides in aging and healthspan. J Clin Invest. 2022; 132:e158449.
144. Cobb LJ, Lee C, Xiao J, Yen K, Wong RG, Nakamura HK, et al. Naturally occurring mitochondrial-derived peptides are age-dependent regulators of apoptosis, insulin sensitivity, and inflammatory markers. Aging (Albany NY). 2016; 8:796–809.
145. Miller B, Kim SJ, Kumagai H, Mehta HH, Xiang W, Liu J, et al. Peptides derived from small mitochondrial open reading frames: genomic, biological, and therapeutic implications. Exp Cell Res. 2020; 393:112056.
146. Kang GM, Min SH, Lee CH, Kim JY, Lim HS, Choi MJ, et al. Mitohormesis in hypothalamic POMC neurons mediates regular exercise-induced high-turnover metabolism. Cell Metab. 2021; 33:334–49.
147. Guo B, Zhai D, Cabezas E, Welsh K, Nouraini S, Satterthwait AC, et al. Humanin peptide suppresses apoptosis by interfering with Bax activation. Nature. 2003; 423:456–61.
148. Cai H, Liu Y, Men H, Zheng Y. Protective mechanism of humanin against oxidative stress in aging-related cardiovascular diseases. Front Endocrinol (Lausanne). 2021; 12:683151.
149. Kim KH, Son JM, Benayoun BA, Lee C. The mitochondrialencoded peptide MOTS-c translocates to the nucleus to regulate nuclear gene expression in response to metabolic stress. Cell Metab. 2018; 28:516–24.
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr