CLINICAL PHARMACOLOGY / STATE OF THE ART PAPER
Impact of rapamycin on longevity: updated insights
More details
Hide details
1
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
2
Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
3
Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
4
Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
5
Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
6
Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
7
Liverpool Centre for Cardiovascular Science at University of Liverpool (LCCS), Liverpool, UK
8
Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
9
Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
10
Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
These authors had equal contribution to this work
Submission date: 2023-11-21
Final revision date: 2024-07-02
Acceptance date: 2024-07-24
Online publication date: 2024-07-25
Corresponding author
Maciej Banach
Department of Preventive
Cardiology and Lipidology
Medical University of Lodz
Rzgowska 281/289
93-228 Lodz, Poland
Phone: +48422711124
Amirhossein Sahebkar
Biotechnology Research
Center
Pharmaceutical Technology
Institute
Mashhad University
of Medical Sciences
Mashhad, Iran
KEYWORDS
TOPICS
ABSTRACT
The gradual accumulation of varying detrimental alterations during the aging process within cells and tissues contributes to a progressive decline in their functionality, which may ultimately result in death. The licensed mammalian target of rapamycin (mTOR) inhibitor rapamycin, also known as sirolimus, has recently become a promising option for anti-aging applications. Through in vitro and in vivo assessments, numerous scientific reports have illustrated diverse biochemical and clinical aspects of rapamycin’s pharmacological effects in ameliorating aging-related changes and expanding longevity. Nevertheless, its clinical application has been impeded by severe adverse effects, which might be addressed by implementing an appropriate therapeutic regimen. In this regard, integrating updated insights and uncovering essential benefits and drawbacks of rapamycin as a geroprotective drug are critical for conducting further preclinical research and well-organized clinical trials, and facilitating translation to clinical practice. The present review highlights the recent findings on the role of rapamycin in improving organ health and postponing aging-related processes.
REFERENCES (245)
1.
Partridge L, Deelen J, Slagboom PE. Facing up to the global challenges of ageing. Nature 2018; 561: 45-56.
2.
Fang EF, Scheibye-Knudsen M, Jahn HJ, et al. A research agenda for aging in China in the 21st century. Ageing Res Rev 2015; 24: 197-205.
3.
Moskalev A, Guvatova Z, Lopes IDA, et al. Targeting aging mechanisms: pharmacological perspectives. Trends Endocrinol Metabol 2022; 33: 266-80.
4.
Papadopoli D, Boulay K, Kazak L, et al. mTOR as a central regulator of lifespan and aging. F1000Res 2019; 8: F1000 Faculty Rev-998.
5.
Weichhart T. mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology 2018; 64: 127-34.
6.
Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature 2013; 493: 338-45.
7.
Blagosklonny MV. Rapamycin for longevity: opinion article. Aging (Albany NY) 2019; 11: 8048.
8.
Wang D, Eisen HJ. Mechanistic target of rapamycin (mTOR) inhibitors. Handb Exp Pharmacol 2022; 272: 53-72.
9.
Zuo R, Wang Y, Li J, et al. Rapamycin induced autophagy inhibits inflammation-mediated endplate degeneration by enhancing Nrf2/Keap1 signaling of cartilage endplate stem cells. Stem Cells 2019; 37: 828-40.
10.
Juricic P, Lu YX, Leech T, et al. Long-lasting geroprotection from brief rapamycin treatment in early adulthood by persistently increased intestinal autophagy. Nature Aging 2022; 2: 824-36.
11.
Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 2022; 18: 243-58.
12.
Sorrenti V, Benedetti F, Buriani A, et al. Immunomodulatory and antiaging mechanisms of resveratrol, rapamycin, and metformin: focus on mTOR and AMPK signaling networks. Pharmaceuticals 2022; 15: 912.
13.
Lamming DW, Ye L, Katajisto P, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science 2012; 335: 1638-43.
14.
Kim J, Guan KL. mTOR as a central hub of nutrient signalling and cell growth. Nat Cell Biol 2019; 21: 63-71.
15.
Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 2011; 12: 21-35.
16.
Jhanwar-Uniyal M, Wainwright JV, Mohan AL, et al. Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regulation 2019; 72: 51-62.
17.
Dibble CC, Manning BD. Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat Cell Biol 2013; 15: 555-64.
18.
Groenewoud MJ, Zwartkruis FJ. Rheb and Rags come together at the lysosome to activate mTORC1. Biochem Soc Transact 2013; 41: 951-5.
19.
Shen K, Huang RK, Brignole EJ, et al. Architecture of the human GATOR1 and GATOR1–Rag GTPases complexes. Nature 2018; 556: 64-9.
20.
Chantranupong L, Wolfson RL, Orozco JM, et al. The Sestrins interact with GATOR2 to negatively regulate the amino-acid-sensing pathway upstream of mTORC1. Cell Rep 2014; 9: 1-8.
21.
Wolfson RL, Chantranupong L, Saxton RA, et al. Sestrin2 is a leucine sensor for the mTORC1 pathway. Science 2016; 351: 43-8.
22.
Saxton RA, Chantranupong L, Knockenhauer KE, Schwartz TU, Sabatini DM. Mechanism of arginine sensing by CASTOR1 upstream of mTORC1. Nature 2016; 536: 229-33.
23.
Wang S, Tsun ZY, Wolfson RL, et al. Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1. Science 2015; 347: 188-94.
24.
Tsun ZY, Bar-Peled L, Chantranupong L, et al. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol Cell 2013; 52: 495-505.
25.
Dibble CC, Cantley LC. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol 2015; 25: 545-55.
26.
Easton JB, Kurmasheva RT, Houghton PJ. IRS-1: auditing the effectiveness of mTOR inhibitors. Cancer Cell 2006; 9: 153-5.
27.
Inoki K, Ouyang H, Zhu T, et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 2006; 126: 955-68.
28.
Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosisand cancer pathogenesis. Cell 2005; 121: 179-93.
29.
Liu Y, Cao GF, Xue J, et al. Tumor necrosis factor-alpha (TNF-)-mediated in vitro human retinal pigment epithelial (RPE) cell migration mainly requires Akt/mTOR complex 1 (mTORC1), but not mTOR complex 2 (mTORC2) signaling. Eur J Cell Biol 2012; 91: 728-37.
30.
Jastrzebski K, Hannan KM, Tchoubrieva EB, Hannan RD, Pearson RB. Coordinate regulation of ribosome biogenesis and function by the ribosomal protein S6 kinase, a key mediator of mTOR function. Growth Factors 2007; 25: 209-26.
31.
Haar EV, Lee S, Bandhakavi S, Griffin TJ, Kim DH. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol 2007; 9: 316-23.
32.
Van Nostrand JL, Hellberg K, Luo EC, et al. AMPK regulation of Raptor and TSC2 mediate metformin effects on transcriptional control of anabolism and inflammation. Genes Developm 2020; 34: 1330-44.
33.
Shang C, Zhou H, Liu W, Shen T, Luo Y, Huang S. Iron chelation inhibits mTORC1 signaling involving activation of AMPK and REDD1/Bnip3 pathways. Oncogene 2020; 39: 5201-13.
34.
Ma Y, Vassetzky Y, Dokudovskaya S. mTORC1 pathway in DNA damage response. Biochim Biophys Acta Mol Cell Res 2018; 1865: 1293-311.
35.
Ding B, Parmigiani A, Divakaruni AS, Archer K, Murphy AN, Budanov AV. Sestrin2 is induced by glucose starvation via the unfolded protein response and protects cells from non-canonical necroptotic cell death. Sci Rep 2016; 6: 22538.
36.
Yang M, Lu Y, Piao W, Jin H. The translational regulation in mTOR pathway. Biomolecules 2022; 12: 802.
37.
Dorrello NV, Peschiaroli A, Guardavaccaro D, Colburn NH, Sherman NE, Pagano M. S6K1-and ßTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science 2006; 314: 467-71.
38.
Ma XM, Yoon SO, Richardson CJ, Jülich K, Blenis J. SKAR links pre-mRNA splicing to mTOR/S6K1-mediated enhanced translation efficiency of spliced mRNAs. Cell 2008; 133: 303-13.
39.
Jiao L, Liu Y, Yu XY, et al. Ribosome biogenesis in disease: new players and therapeutic targets. Signal Transd Targeted Ther 2023; 8: 15.
40.
Mayer C, Grummt I. Ribosome biogenesis and cell growth: mTOR coordinates transcription by all three classes of nuclear RNA polymerases. Oncogene 2006; 25: 6384-91.
41.
Lee G, Zheng Y, Cho S, et al. Post-transcriptional regulation of de novo lipogenesis by mTORC1-S6K1-SRPK2 signaling. Cell 2017; 171: 1545-58.e18.
42.
Peterson TR, Sengupta SS, Harris TE, et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 2011; 146: 408-20.
43.
Guo Z, Cheng X, Feng X, et al. The mTORC1/4EBP1/PPAR axis mediates insulin-induced lipogenesis by regulating lipogenic gene expression in bovine mammary epithelial cells. J Agricul Food Chem 2019; 67: 6007-18.
44.
Ben-Sahra I, Hoxhaj G, Ricoult SJ, Asara JM, Manning BD. mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 2016; 351: 728-33.
45.
Robitaille AM, Christen S, Shimobayashi M, et al. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 2013; 339: 1320-3.
46.
Dodd KM, Yang J, Shen MH, Sampson JR, Tee AR. mTORC1 drives HIF-1 and VEGF-A signalling via multiple mechanisms involving 4E-BP1, S6K1 and STAT3. Oncogene 2015; 34: 2239-50.
47.
Valvezan AJ, Manning BD. Molecular logic of mTORC1 signalling as a metabolic rheostat. Nature Metabol 2019; 1: 321-33.
48.
de la Cruz López KG, Toledo Guzmán ME, Sánchez EO, García Carrancá A. mTORC1 as a regulator of mitochondrial functions and a therapeutic target in cancer. Front Oncol 2019; 9: 1373.
49.
Morita M, Gravel SP, Chenard V, et al. mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation. Cell Metabol 2013; 18: 698-711.
50.
Hosokawa N, Hara T, Kaizuka T, et al. Nutrient-dependent mTORC1 association with the ULK1–Atg13–FIP200 complex required for autophagy. Mol Biol Cell 2009; 20: 1981-91.
51.
Kim YM, Jung CH, Seo M, et al. mTORC1 phosphorylates UVRAG to negatively regulate autophagosome and endosome maturation. Mol Cell 2015; 57: 207-18.
52.
Nnah IC, Wang B, Saqcena C, et al. TFEB-driven endocytosis coordinates MTORC1 signaling and autophagy. Autophagy 2019; 15: 151-64.
53.
Martina JA, Diab HI, Brady OA, Puertollano R. TFEB and TFE 3 are novel components of the integrated stress response. EMBO J 2016; 35: 479-95.
54.
Odle RI, Walker SA, Oxley D, et al. An mTORC1-to-CDK1 switch maintains autophagy suppression during mitosis. Mol Cell 2020; 77: 228-40.e7.
55.
Liu P, Gan W, Chin YR, et al. PtdIns (3, 4, 5) P3-dependent activation of the mTORC2 Kinase ComplexPtdIns (3, 4, 5) P3 activates mTORC2 by binding the SIN1-PH domain. Cancer Discov 2015; 5: 1194-209.
56.
Humphrey SJ, Yang G, Yang P, et al. Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metabol 2013; 17: 1009-20.
57.
Ebner M, Sinkovics B, Szczygieł M, Ribeiro DW, Yudushkin I. Localization of mTORC2 activity inside cells. J Cell Biol 2017; 216: 343-53.
58.
Castel P, Dharmaiah S, Sale MJ, et al. RAS interaction with Sin1 is dispensable for mTORC2 assembly and activity. Proc Natl Acad Sci 2021; 118: e2103261118.
59.
Cahigas GM, Charest P. Determining the role of the Ras family GTPase Rap1 in regulating mTORC2 activity and function in cell migration. FASEB J 2022; 36.
60.
Kovalski JR, Bhaduri A, Zehnder AM, et al. The functional proximal proteome of oncogenic Ras includes mTORC2. Mol Cell 2019; 73: 830-44.e12.
61.
Kazyken D, Magnuson B, Bodur C, et al. AMPK directly activates mTORC2 to promote cell survival during acute energetic stress. Sci Signal 2019; 12: eaav3249.
62.
Yuan HX, Guan KL. The SIN1-PH domain connects mTORC2 to PI3K. Cancer Discov 2015; 5: 1127-9.
63.
Sarbassov DD, Ali SM, Kim DH, et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 2004; 14: 1296-302.
64.
Larsson C. Protein kinase C and the regulation of the actin cytoskeleton. Cell Signal 2006; 18: 276-84.
65.
Kim LC, Cook RS, Chen J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2017; 36: 2191-201.
66.
Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005; 307: 1098-101.
67.
Hoxhaj G, Ben-Sahra I, Lockwood SE, et al. Direct stimulation of NADP+ synthesis through Akt-mediated phosphorylation of NAD kinase. Science 2019; 363: 1088-92.
68.
Yadav RK, Chauhan AS, Zhuang L, Gan B. FoxO transcription factors in cancer metabolism. Semin Cancer Biol 2018; 50: 65-76.
69.
Maurer U, Preiss F, Brauns-Schubert P, Schlicher L, Charvet C. GSK-3–at the crossroads of cell death and survival. J Cell Sci 2014; 127: 1369-78.
70.
Guertin DA, Stevens DM, Thoreen CC, et al. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKC, but not S6K1. Develop Cell 2006; 11: 859-71.
71.
Yu Y, Yoon SO, Poulogiannis G, et al. Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling. Science 2011; 332: 1322-6.
72.
Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol 2002; 4: 648-57.
73.
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 2020; 21: 183-203.
74.
Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell Metabol 2014; 19: 373-9.
75.
Klaips CL, Jayaraj GG, Hartl FU. Pathways of cellular proteostasis in aging and disease. J Cell Biol 2018; 217: 51-63.
76.
Labbadia J, Morimoto RI. The biology of proteostasis in aging and disease. Ann Rev Biochem 2015; 84: 435-64.
77.
Conn CS, Qian SB. Nutrient signaling in protein homeostasis: an increase in quantity at the expense of quality. Sci Signal 2013; 6: ra24-ra.
78.
Kaeberlein M, Kennedy BK. Hot topics in aging research: protein translation and TOR signaling, 2010. Aging Cell 2011; 10: 185-90.
79.
Blackwell TK, Sewell AK, Wu Z, Han M. TOR signaling in Caenorhabditis elegans development, metabolism, and aging. Genetics 2019; 213: 329-60.
80.
Wilson KA, Bar S, Kapahi P. Haste makes waste: the significance of translation fidelity for development and longevity. Mol Cell 2021; 81: 3675-6.
81.
Martinez-Miguel VE, Lujan C, Espie T, et al. Increased fidelity of protein synthesis extends lifespan. Cell Metabol 2021; 33: 2288-300.e12.
82.
Xie J, de Souza Alves V, von der Haar T, et al. Regulation of the elongation phase of protein synthesis enhances translation accuracy and modulates lifespan. Curr Biol 2019; 29: 737-49.e5.
83.
Yee Z, Lim SHY, Ng LF, Gruber J. Inhibition of mTOR decreases insoluble proteins burden by reducing translation in C. elegans. Biogerontology 2021; 22: 101-18.
84.
Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci 2008; 105: 17414-9.
85.
Ozkurede U, Kala R, Johnson C, Shen Z, Miller RA, Garcia GG. Cap-independent mRNA translation is upregulated in long-lived endocrine mutant mice. J Mol Endocrinol 2019; 63: 123-38.
86.
Roux PP, Topisirovic I. Signaling pathways involved in the regulation of mRNA translation. Mol Cell Biol 2018; 38: e00070-18.
87.
Lacerda R, Menezes J, Romão L. More than just scanning: the importance of cap-independent mRNA translation initiation for cellular stress response and cancer. Cell Mol Life Sci 2017; 74: 1659-80.
88.
Lence T, Soller M, Roignant JY. A fly view on the roles and mechanisms of the m6A mRNA modification and its players. RNA Biol 2017; 14: 1232-40.
89.
Shen Z, Hinson A, Miller RA, Garcia GG. Cap-independent translation: a shared mechanism for lifespan extension by rapamycin, acarbose, and 17-estradiol. Aging Cell 2021; 20: e13345.
90.
Statzer C, Meng J, Venz R, et al. ATF-4 and hydrogen sulfide signalling mediate longevity in response to inhibition of translation or mTORC1. Nat Commun 2022; 13: 967.
91.
Mittal N, Guimaraes JC, Gross T, et al. The Gcn4 transcription factor reduces protein synthesis capacity and extends yeast lifespan. Nat Commun 2017; 8: 457.
92.
Laberge RM, Sun Y, Orjalo AV, et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol 2015; 17: 1049-61.
93.
Herranz N, Gallage S, Mellone M, et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol 2015; 17: 1205-17.
94.
Canino C, Mori F, Cambria A, et al. SASP mediates chemoresistance and tumor-initiating-activity of mesothelioma cells. Oncogene 2012; 31: 3148-63.
95.
Sun L, Yan Y, Lv H, et al. Rapamycin targets STAT3 and impacts c-Myc to suppress tumor growth. Cell Chem Biol 2022; 29: 373-85.e6.
96.
Aman Y, Schmauck-Medina T, Hansen M, et al. Autophagy in healthy aging and disease. Nature Aging 2021; 1: 634-50.
97.
Schiattarella GG, Hill JA. Therapeutic targeting of autophagy in cardiovascular disease. J Mol Cell Cardiol 2016; 95: 86-93.
98.
Hansen M, Rubinsztein DC, Walker DW. Autophagy as a promoter of longevity: insights from model organisms. Nat Rev Mol Cell Biol 2018; 19: 579-93.
99.
Wong SQ, Kumar AV, Mills J, Lapierre LR. Autophagy in aging and longevity. Hum Genet 2020; 139: 277-90.
100.
Kaushik S, Tasset I, Arias E, et al. Autophagy and the hallmarks of aging. Ageing Res Rev 2021; 72:.
101.
Hansen M, Chandra A, Mitic LL, Onken B, Driscoll M, Kenyon C. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genet 2008; 4: e24.
102.
Alvers AL, Wood MS, Hu D, et al. Autophagy is required for extension of yeast chronological life span by rapamycin. Autophagy 2009; 5: 847-9.
103.
García-Prat L, Martínez-Vicente M, Perdiguero E, et al. Autophagy maintains stemness by preventing senescence. Nature 2016; 529: 37-42.
104.
Li W, Wang H, Tan Y, Wang Y, Yu S, Li Z. Reducing lipofuscin accumulation and cardiomyocytic senescence of aging heart by enhancing autophagy. Exp Cell Res 2021; 403: 112585.
105.
Gao G, Chen W, Yan M, et al. Rapamycin regulates the balance between cardiomyocyte apoptosis and autophagy in chronic heart failure by inhibiting mTOR signaling. Int J Mol Med 2020; 45: 195-209.
106.
Zhang C, Liu A, Su G, Chen Y. Effect of rapamycin on the level of autophagy in rats with early heart failure. J Cell Biochem 2019; 120: 4065-70.
107.
Pulakat L, Chen HH. Pro-senescence and anti-senescence mechanisms of cardiovascular aging: cardiac microRNA regulation of longevity drug-induced autophagy. Front Pharmacol 2020; 11: 774.
108.
Munasinghe PE, Riu F, Dixit P, et al. Type-2 diabetes increases autophagy in the human heart through promotion of Beclin-1 mediated pathway. Int J Cardiol 2016; 202: 13-20.
109.
Young AR, Narita M, Ferreira M, et al. Autophagy mediates the mitotic senescence transition. Genes Develop 2009; 23: 798-803.
110.
Liu CY, Zhang YH, Li RB, et al. LncRNA CAIF inhibits autophagy and attenuates myocardial infarction by blocking p53-mediated myocardin transcription. Nat Commun 2018; 9: 29.
111.
Li J, Zhang D, Wiersma M, Brundel BJ. Role of autophagy in proteostasis: friend and foe in cardiac diseases. Cells 2018; 7: 279.
112.
Shi B, Ma M, Zheng Y, Pan Y, Lin X. mTOR and Beclin1: two key autophagy-related molecules and their roles in myocardial ischemia/reperfusion injury. J Cell Physiol 2019; 234: 12562-8.
113.
Zhao J, Zhang J, Liu Q, et al. Hongjingtian injection protects against myocardial ischemia reperfusion-induced apoptosis by blocking ROS induced autophagic-flux. Biomed Pharmacother 2021; 135: 111205.
114.
Huang G, Lu X, Duan Z, et al. PCSK9 knockdown can improve myocardial ischemia/reperfusion injury by inhibiting autophagy. Cardiovasc Toxicol 2022; 22: 951-61.
115.
Kim CK, Sachdev PS, Braidy N. Recent neurotherapeutic strategies to promote healthy brain aging: are we there yet? Aging Dis 2022; 13: 175.
116.
Marzoog BA. Autophagy as an anti-senescent in aging neurocytes. Curr Mol Med 2023; 24: 182-90.
117.
Singh AK, Singh S, Tripathi VK, Bissoyi A, Garg G, Rizvi SI. Rapamycin confers neuroprotection against aging-induced oxidative stress, mitochondrial dysfunction, and neurodegeneration in old rats through activation of autophagy. Rejuvenation Res 2019; 22: 60-70.
118.
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78: 101636.
119.
Yang N, Liu X, Niu X, et al. Activation of autophagy ameliorates age-related neurogenesis decline and neurodysfunction in adult mice. Stem Cell Rev Rep 2022; 18: 626-41.
120.
Norwitz NG, Querfurth H. mTOR mysteries: nuances and questions about the mechanistic target of rapamycin in neurodegeneration. Front Neurosci 2020; 14: 775.
121.
Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med 2013; 19: 983-97.
122.
Selvarani R, Mohammed S, Richardson A. Effect of rapamycin on aging and age-related diseases – past and future. Geroscience 2021; 43: 1135-58.
123.
Kakoty V, Yang CH, Kumari S, Dubey SK, Taliyan R. Neuroprotective effect of lentivirus-mediated FGF21 gene delivery in experimental Alzheimer’s disease is augmented when concerted with rapamycin. Mol Neurobiol 2022; 59: 2659-77.
124.
Hashemian S, Shojaei M, Radbakhsh S, et al. The effects of oral trehalose on glycaemia, inflammation, and quality of life in patients with type 2 diabetes: a pilot randomized controlled trial. Arch Med Sci 2023; 19: 1693-700.
125.
Hosseinpour-Moghaddam K, Caraglia M, Sahebkar A. Autophagy induction by trehalose: molecular mechanisms and therapeutic impacts. J Cell Physiol 2018; 233: 6524-43.
126.
Khalifeh M, Barreto G, Sahebkar A. Therapeutic potential of trehalose in neurodegenerative diseases: the knowns and unknowns. Neural Regen Res 2021; 16: 2026-7.
127.
Khalifeh M, Barreto GE, Sahebkar A. Trehalose as a promising therapeutic candidate for the treatment of Parkinson’s disease. Br J Pharmacol 2019; 176: 1173-89.
128.
Ganjali S, Mansouri A, Reiner Ž, et al. The effect of trehalose administration on the serum expression levels of microRNAs associated with lipid metabolism and the autophagy process in patients with myocardial infarction – post-hoc analysis of the IR-TREAT trial. Arch Med Sci 2023; doi: 10.5114/aoms/170159.
129.
Ganjali S, Jamialahmadi T, Abbasifard M, et al. Trehalose-induced alterations in serum expression levels of microRNAs associated with vascular inflammation in patients with coronary artery disease – the pilot results from the randomized controlled trial. Arch Med Sci 2022; doi: 10.5114/aoms/154987.
130.
Pupyshev AB, Tikhonova MA, Akopyan AA, Tenditnik MV, Dubrovina NI, Korolenko TA. Therapeutic activation of autophagy by combined treatment with rapamycin and trehalose in a mouse MPTP-induced model of Parkinson’s disease. Pharmacol Biochem Behav 2019; 177: 1-11.
131.
Liu T, Wang P, Yin H, et al. Rapamycin reverses ferroptosis by increasing autophagy in MPTP/MPP+-induced models of Parkinson’s disease. Neural Regen Res 2023; 18: 2514-9.
132.
Carosi JM, Sargeant TJ. Rapamycin and Alzheimer disease: a double-edged sword? Autophagy 2019; 15: 1460-2.
133.
Russell OM, Gorman GS, Lightowlers RN, Turnbull DM. Mitochondrial diseases: hope for the future. Cell 2020; 181: 168-88.
134.
Akbari M, Kirkwood TB, Bohr VA. Mitochondria in the signaling pathways that control longevity and health span. Ageing Res Rev 2019; 54: 100940.
135.
Leontieva OV, Blagosklonny MV. M (o) TOR of pseudo-hypoxic state in aging: rapamycin to the rescue. Cell Cycle 2014; 13: 509-15.
136.
Amjad S, Nisar S, Bhat AA, et al. Role of NAD+ in regulating cellular and metabolic signaling pathways. Mol Metab 2021; 49: 101195.
137.
Gomes AP, Price NL, Ling AJ, et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 2013; 155: 1624-38.
138.
Zhang Z, Xu HN, Li S, et al. Rapamycin maintains NAD+/NADH redox homeostasis in muscle cells. Aging 2020; 12: 17786.
139.
Chiao YA, Kolwicz SC, Basisty N, et al. Rapamycin transiently induces mitochondrial remodeling to reprogram energy metabolism in old hearts. Aging 2016; 8: 314.
140.
Huwatibieke B, Yin W, Liu L, et al. Mammalian target of rapamycin signaling pathway regulates mitochondrial quality control of brown adipocytes in mice. Front Physiol 2021; 12: 63852.
141.
Bielas J, Herbst A, Widjaja K, et al. Long term rapamycin treatment improves mitochondrial DNA quality in aging mice. Exp Gerontol 2018; 106: 125-31.
142.
Martínez-Cisuelo V, Gómez J, García-Junceda I, et al. Rapamycin reverses age-related increases in mitochondrial ROS production at complex I, oxidative stress, accumulation of mtDNA fragments inside nuclear DNA, and lipofuscin level, and increases autophagy, in the liver of middle-aged mice. Exp Gerontol 2016; 83: 130-8.
143.
Siegmund SE, Yang H, Sharma R, et al. Low-dose rapamycin extends lifespan in a mouse model of mtDNA depletion syndrome. Human Mol Genet 2017; 26: 4588-605.
144.
Winans T, Oaks Z, Choudhary G, et al. mTOR-dependent loss of PON1 secretion and antiphospholipid autoantibody production underlie autoimmunity-mediated cirrhosis in transaldolase deficiency. J Autoimmun 2023; 140: 103112.
145.
Oaks Z, Winans T, Caza T, et al. Mitochondrial dysfunction in the liver and antiphospholipid antibody production precede disease onset and respond to rapamycin in lupus-prone mice. Arthritis Rheumatol 2016; 68: 2728-39.
146.
Oaks Z, Patel A, Huang N, et al. Cytosolic aldose metabolism contributes to progression from cirrhosis to hepatocarcinogenesis. Nat Metabol 2023; 5: 41-60.
147.
Mahalakshmi R, Priyanga J, Bhakta-Guha D, Guha G. Hormetic alteration of mTOR-mitochondria association: an approach to mitigate cellular aging. Curr Opin Environ Sci Health 2022; 29:100387.
148.
Barriocanal-Casado E, Hidalgo-Gutiérrez A, Raimundo N, et al. Rapamycin administration is not a valid therapeutic strategy for every case of mitochondrial disease. EBioMedicine 2019; 42: 511-23.
149.
Kallijärvi J, Fellman V. Rapamycin –“One size does not fit all”. EBioMedicine 2019; 42: 30-1.
150.
Yousefzadeh MJ, Flores RR, Zhu Y, et al. An aged immune system drives senescence and ageing of solid organs. Nature 2021; 594: 100-5.
151.
Baylis D, Bartlett DB, Patel HP, Roberts HC. Understanding how we age: insights into inflammaging. Longev Healthspan 2013; 2: 8.
152.
Nazari N, Jafari F, Ghalamfarsa G, et al. The emerging role of microRNA in regulating the mTOR signaling pathway in immune and inflammatory responses. Immunol Cell Biol 2021; 99: 814-32.
153.
Zhang P, Catterson JH, Grönke S, Partridge L. Inhibition of S6K lowers age-related inflammation and immunosenescence and increases lifespan through the endolysosomal system. bioRxiv 2022; 2022.08. 25.505264.
154.
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol 2020; 42: 559-72.
155.
Barbé-Tuana F, Funchal G, Schmitz CRR, Maurmann RM, Bauer ME. The interplay between immunosenescence and age-related diseases. Semin Immunopathol 2020; 42: 545-57.
156.
Suto T, Karonitsch T. The immunobiology of mTOR in autoimmunity. J Autoimmun 2020; 110: 102373.
157.
Baroja-Mazo A, Revilla-Nuin B, Ramírez P, Pons JA. Immunosuppressive potency of mechanistic target of rapamycin inhibitors in solid-organ transplantation. World J Transplantation 2016; 6: 183-92.
158.
Zhang M, Chong KK, Chen Z, et al. Rapamycin improves Graves’ orbitopathy by suppressing CD4+ cytotoxic T lymphocytes. JCI Insight 2023; 8: e160377.
159.
Lai ZW, Kelly R, Winans T, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet 2018; 391: 1186-96.
160.
Hu X, Zhang H, Li X, Li Y, Chen Z. Activation of mTORC1 in fibroblasts accelerates wound healing and induces fibrosis in mice. Wound Repair Regen 2020; 28: 6-15.
161.
Namba DR, Ma G, Samad I, et al. Rapamycin inhibits human laryngotracheal stenosis-derived fibroblast proliferation, metabolism, and function in vitro. Otolaryngol Head Neck Surg 2015; 152: 881-8.
162.
O’Shea AE, Valdera FA, Ensley D, et al. Immunologic and dose dependent effects of rapamycin and its evolving role in chemoprevention. Clin Immunol 2022; 245: 109095.
163.
Boada C, Zinger A, Tsao C, et al. Rapamycin-loaded biomimetic nanoparticles reverse vascular inflammation. Circ Res 2020; 126: 25-37.
164.
Wang J, Chai L, Lu Y, Lu H, Liu Y, Zhang Y. Attenuation of mTOR signaling is the major response element in the rescue pathway of chronic kidney disease in rats. Neuroimmunomodulation 2020; 27: 9-18.
165.
Bhatt K, Bhagavathula M, Verma S, et al. Rapamycin modulates pulmonary pathology in a murine model of Mycobacterium tuberculosis infection. Dis Models Mechanisms 2021; 14: dmm049018.
166.
Wink L, Miller RA, Garcia GG. Rapamycin, acarbose and 17-estradiol share common mechanisms regulating the MAPK pathways involved in intracellular signaling and inflammation. Immun Ageing 2022; 19: 8.
167.
Zwaans V, Mohr F, Remes A, et al. The anti-inflammatory capacity of rapamycin in aortic vascular smooth muscle cells from marfan syndrome mice. Thorac Cardiovasc Surg 2023; 71: DGTHG-V13.
168.
Ge C, Ma C, Cui J, et al. Rapamycin suppresses inflammation and increases the interaction between p65 and IB in rapamycin-induced fatty livers. PLoS One 2023; 18: e0281888.
169.
Dai J, Jiang C, Chen H, Chai Y. Rapamycin attenuates high glucose-induced inflammation through modulation of mTOR/NF-B pathways in macrophages. Front Pharmacol 2019; 10: 1292.
170.
Jia X, Cao B, An Y, Zhang X, Wang C. Rapamycin ameliorates lipopolysaccharide-induced acute lung injury by inhibiting IL-1 and IL-18 production. Int Immunopharmacol 2019; 67: 211-9.
171.
Correia-Melo C, Birch J, Fielder E, et al. Rapamycin improves healthspan but not inflammaging in nfb1−/− mice. Aging Cell 2019; 18: e12882.
172.
Phillips EJ, Simons MJ. Rapamycin not dietary restriction improves resilience against pathogens: a meta-analysis. GeroScience 2023; 45: 1263-70.
173.
Yende S, Tuomanen EI, Wunderink R, et al. Preinfection systemic inflammatory markers and risk of hospitalization due to pneumonia. Am J Respir Crit Care Med 2005; 172: 1440-6.
174.
MacNee W. Accelerated lung aging: a novel pathogenic mechanism of chronic obstructive pulmonary disease (COPD). Biochem Soc Trans 2009; 37: 819-23.
175.
Infante AJ, McCullers JA, Orihuela CJ. Mechanisms of predisposition to pneumonia: infants, the elderly, and viral infections. In: Streptococcus Pneumoniae. Molecular Mechanisms of Host-Pathogen Interactions. Brown J, Hammerschmidt S, Orihuela C (eds.). Elsevier 2015: 363-82.
176.
Mu W, Rezek V, Martin H, et al. Autophagy inducer rapamycin treatment reduces IFN-I-mediated Inflammation and improves anti-HIV-1 T cell response in vivo. JCI Insight 2022; 7: e159136.
177.
Ghadimi M, Mohammadpour Z, Dashti-Khavidaki S, Milajerdi A. m-TOR inhibitors and risk of Pneumocystis pneumonia after solid organ transplantation: a systematic review and meta-analysis. Eur J Clin Pharmacol 2019; 75: 1471-80.
178.
Dominguez J, Mahalati K, Kiberd B, McAlister VC, MacDonald AS. Conversion to rapamycin immunosuppression in renal transplant recipients: report of an initial experience. Transplantation 2000; 70: 1244-7.
179.
Nam JH. Rapamycin: could it enhance vaccine efficacy? Exp Rev Vaccines 2009; 8: 1535-9.
180.
Moraschi BF, Noronha IH, Ferreira CP, et al. Rapamycin improves the response of effector and memory CD8+ T cells induced by immunization with ASP2 of Trypanosoma cruzi. Front Cell Infect Microbiol 2021; 11: 676183.
181.
Jagannath C, Bakhru P. Rapamycin-induced enhancement of vaccine efficacy in mice. Methods Mol Biol 2012; 821: 295-303.
182.
Jagannath C, Lindsey DR, Dhandayuthapani S, Xu Y, Hunter Jr RL, Eissa NT. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Nat Med 2009; 15: 267-76.
183.
Li Q, Rao R, Vazzana J, et al. Regulating mammalian target of rapamycin to tune vaccination-induced CD8+ T cell responses for tumor immunity. J Immunol 2012; 188: 3080-7.
184.
Amiel E, Everts B, Freitas TC, et al. Inhibition of mechanistic target of rapamycin promotes dendritic cell activation and enhances therapeutic autologous vaccination in mice. J Immunol 2012; 189: 2151-8.
185.
Sun IH, Oh MH, Zhao L, et al. mTOR complex 1 signaling regulates the generation and function of central and effector Foxp3+ regulatory T cells. J Immunol 2018; 201: 481-92.
186.
Chaoul N, Fayolle C, Desrues B, et al. Rapamycin impairs antitumor CD8+ T-cell responses and vaccine-induced tumor eradication. Cancer Res 2015; 75: 3279-91.
187.
Bader JE, Voss K, Rathmell JC. Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol Cell 2020; 78: 1019-33.
188.
He D, Wu H, Xiang J, et al. Gut stem cell aging is driven by mTORC1 via a p38 MAPK-p53 pathway. Nat Commun 2020; 11: 37.
189.
Feng R, Wu S, Li R, et al. mTORC1-induced bone marrow-derived mesenchymal stem cell exhaustion contributes to the bone abnormalities in klotho-deficient mice of premature aging aging. Stem Cells Dev 2023; 32: 331-45.
190.
Kawakami Y, Hambright WS, Takayama K, et al. Rapamycin rescues age-related changes in muscle-derived stem/progenitor cells from progeroid mice. Mol Ther Methods Clin Dev 2019; 14: 64-76.
191.
Cao Y, Chen W, Lei Q, et al. The transplantation of rapamycin-treated senescent human mesenchymal stem cells with enhanced proangiogenic activity promotes neovascularization and ischemic limb salvage in mice. Acta Pharmacol Sin 2022; 43: 2885-94.
192.
Nie D, Zhang J, Zhou Y, et al. Rapamycin treatment of tendon stem/progenitor cells reduces cellular senescence by upregulating autophagy. Stem Cells Int 2021; 2021: 6638249.
193.
Hu C, Zhao L, Wu D, Li L. Modulating autophagy in mesenchymal stem cells effectively protects against hypoxia-or ischemia-induced injury. Stem Cell Res Ther 2019; 10: 120.
194.
Ceccariglia S, Cargnoni A, Silini AR, Parolini O. Autophagy: a potential key contributor to the therapeutic action of mesenchymal stem cells. Autophagy 2020; 16: 28-37.
195.
He J, Liu J, Huang Y, Tang X, Xiao H, Hu Z. Oxidative stress, inflammation, and autophagy: potential targets of mesenchymal stem cells-based therapies in ischemic stroke. Front Neurosci 2021; 15: 641157.
196.
Cen S, Wang P, Xie Z, et al. Autophagy enhances mesenchymal stem cell-mediated CD4+ T cell migration and differentiation through CXCL8 and TGF-1. Stem Cell Res Ther 2019; 10: 265.
197.
Rodolfo C, Di Bartolomeo S, Cecconi F. Autophagy in stem and progenitor cells. Cell Mol Life Sci 2016; 73: 475-96.
198.
Li Z, Wang Y, Wang H, Wu J, Tan Y. Rapamycin-preactivated autophagy enhances survival and differentiation of mesenchymal stem cells after transplantation into infarcted myocardium. Stem Cell Rev Rep 2020; 16: 344-56.
199.
Li M, Lan Y, Gao J, et al. Rapamycin promotes the expansion of myeloid cells by increasing G-CSF expression in mesenchymal stem cells. Front Cell Develop Biol 2022; 10: 779159.
200.
Greenbaum A, Link D. Mechanisms of G-CSF-mediated hematopoietic stem and progenitor mobilization. Leukemia 2011; 25: 211-7.
201.
Xing Y, Liu C, Zhou L, Li Y, Wu D. Osteogenic effects of rapamycin on bone marrow mesenchymal stem cells via inducing autophagy. J Orthop Surg Res 2023; 18: 129.
202.
Pereira-Santos M, Gonçalves-Santos E, Souza MA, et al. Chronic rapamycin pretreatment modulates arginase/inducible nitric oxide synthase balance attenuating aging-dependent susceptibility to Trypanosoma cruzi infection and acute myocarditis. Exp Gerontol 2022; 159: 111676.
203.
Quarles E, Basisty N, Chiao YA, et al. Rapamycin persistently improves cardiac function in aged, male and female mice, even following cessation of treatment. Aging Cell 2020; 19: e13086.
204.
Lesniewski LA, Seals DR, Walker AE, et al. Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell 2017; 16: 17-26.
205.
Yin Z, Guo X, Qi Y, et al. Dietary restriction and rapamycin affect brain aging in mice by attenuating age-related DNA methylation changes. Genes 2022; 13: 699.
206.
Hei C, Zhou Y, Zhang C, et al. Rapamycin ameliorates brain damage and maintains mitochondrial dynamic balance in diabetic rats subjected to middle cerebral artery occlusion. Metab Brain Dis 2023; 38: 409-18.
207.
Lai C, Chen Z, Ding Y, et al. Rapamycin attenuated zinc-induced tau phosphorylation and oxidative stress in rats: involvement of dual mTOR/p70S6K and Nrf2/HO-1 pathways. Front Immunol 2022; 13: 218.
208.
Zhang G, Yin L, Luo Z, et al. Effects and potential mechanisms of rapamycin on MPTP-induced acute Parkinson’s disease in mice. Ann Palliat Med 2021; 10: 2889-97.
209.
Zhang Y, He X, Wu X, et al. Rapamycin upregulates glutamate transporter and IL-6 expression in astrocytes in a mouse model of Parkinson’s disease. Cell Death Dis 2017; 8: e2611.
210.
Fan X, Liang Q, Lian T, et al. Rapamycin preserves gut homeostasis during Drosophila aging. Oncotarget 2015; 6: 35274.
211.
An JY, Kerns KA, Ouellette A, et al. Rapamycin rejuvenates oral health in aging mice. Elife 2020; 9: e54318.
212.
Ding H, Ge G, Tseng Y, Ma Y, Zhang J, Liu J. Hepatic autophagy fluctuates during the development of non-alcoholic fatty liver disease. Ann Hepatol 2020; 19: 516-22.
213.
Houssaini A, Breau M, Kebe K, et al. mTOR pathway activation drives lung cell senescence and emphysema. JCI Insight 2018; 3: e93203.
214.
Mitani A, Ito K, Vuppusetty C, Barnes PJ, Mercado N. Restoration of corticosteroid sensitivity in chronic obstructive pulmonary disease by inhibition of mammalian target of rapamycin. Am J Respir Crit Care Med 2016; 193: 143-53.
215.
Reifsnyder PC, Ryzhov S, Flurkey K, et al. Cardioprotective effects of dietary rapamycin on adult female C57BLKS/J-Leprdb mice. Ann N York Acad Sci 2018; 1418: 106-17.
216.
Zhang S, Zhang Y, Zhang X, et al. Nitrative stress-related autophagic insufficiency participates in hyperhomocysteinemia-induced renal aging. Oxid Med Cell Longev 2020; 2020: 4252047.
217.
Yang Q, Xi Q, Wang M, et al. Rapamycin improves the quality and developmental competence of mice oocytes by promoting DNA damage repair during in vitro maturation. Reprod Biol Endocrinol 2022; 20: 67.
218.
Kuo WW, Baskaran R, Lin JY, et al. Low-dose rapamycin prevents Ang-II-induced toxicity in Leydig cells and testicular dysfunction in hypertensive SHR model. J Biochem Mol Toxicol 2022; 36: e23128.
219.
Orenduff MC, Coleman MF, Glenny EM, et al. Differential effects of calorie restriction and rapamycin on age-related molecular and functional changes in skeletal muscle. Exp Gerontol 2022; 165: 111841.
220.
Tang H, Inoki K, Brooks SV, et al. mTORC1 underlies age-related muscle fiber damage and loss by inducing oxidative stress and catabolism. Aging Cell 2019; 18: e12943.
221.
Arriola Apelo SI, Neuman JC, Baar EL, et al. Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system. Aging Cell 2016; 15: 28-38.
222.
Bitto A, Ito TK, Pineda VV, et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. elife 2016; 5: e16351.
223.
Shindyapina AV, Cho Y, Kaya A, et al. Rapamycin treatment during development extends life span and health span of male mice and Daphnia magna. Sci Adv 2022; 8: eabo5482.
224.
Regan JC, Lu YX, Ureña E, et al. Sexual identity of enterocytes regulates autophagy to determine intestinal health, lifespan and responses to rapamycin. Nat Aging 2022; 2: 1145-58.
225.
Lamming DW, Mihaylova MM, Katajisto P, et al. Depletion of Rictor, an essential protein component of m TORC 2, decreases male lifespan. Aging Cell 2014; 13: 911-7.
226.
Baar EL, Carbajal KA, Ong IM, Lamming DW. Sex-and tissue-specific changes in mTOR signaling with age in C57 BL/6J mice. Aging Cell 2016; 15: 155-66.
227.
Lind MI, Zwoinska MK, Meurling S, Carlsson H, Maklakov AA. Sex-specific tradeoffs with growth and fitness following life-span extension by rapamycin in an outcrossing nematode, Caenorhabditis remanei. J Gerontol A Biomed Sci Med Sci 2016; 71: 882-90.
228.
Strong R, Miller RA, Bogue M, et al. Rapamycin-mediated mouse lifespan extension: late-life dosage regimes with sex-specific effects. Aging Cell 2020; 19: e13269.
229.
Kahan B. Toxicity spectrum of inhibitors of mammalian target of rapamycin in organ transplantation: etiology, pathogenesis and treatment. Exp Opin Drug Safety 2011; 10: 727-49.
230.
Ventura-Aguiar P, Campistol JM, Diekmann F. Safety of mTOR inhibitors in adult solid organ transplantation. Exp Opin Drug Safety 2016; 15: 303-19.
231.
Thibodeau JT, Mishkin JD, Patel PC, et al. Tolerability of sirolimus: a decade of experience at a single cardiac transplant center. Clin Transpl 2013; 27: 945-52.
232.
McCormack FX, Inoue Y, Moss J, et al. Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N Engl J Med 2011; 364: 1595-606
233.
Cabrera López C, Martí T, Catalá V, et al. Effects of rapamycin on angiomyolipomas in patients with tuberous sclerosis. Nefrología 2011; 31: 292-8.
234.
Sadowski K, Sijko K, Domańska-Pakieła D, et al. Antiepileptic effect and safety profile of rapamycin in pediatric patients with tuberous sclerosis complex. Front Neurol 2022; 13: 704978.
235.
Adams DM, Trenor CC, Hammill AM, et al. Efficacy and safety of sirolimus in the treatment of complicated vascular anomalies. Pediatrics 2016; 137: e20153257.
236.
Nadal M, Giraudeau B, Tavernier E, Jonville-Bera AP, Lorette G, Maruani A. Efficacy and safety of mammalian target of rapamycin inhibitors in vascular anomalies: a systematic review. Acta Dermatovenereol 2016; 96: 448-52.
237.
Wen HY, Wang J, Zhang SX, et al. Low-dose sirolimus immunoregulation therapy in patients with active rheumatoid arthritis: a 24-week follow-up of the randomized, open-label, parallel-controlled trial. J Immunol Res 2019; 2019: 7684352.
238.
Stallone G, Infante B, Grandaliano G, Gesualdo L. Management of side effects of sirolimus therapy. Transplantation 2009; 87: S23-6.
239.
Kraig E, Linehan LA, Liang H, et al. A randomized control trial to establish the feasibility and safety of rapamycin treatment in an older human cohort: immunological, physical performance, and cognitive effects. Exp Gerontol 2018; 105: 53-69.
240.
Singh M, Jensen M, Lerman A, et al. Effect of low-dose rapamycin on senescence markers and physical functioning in older adults with coronary artery disease: results of a pilot study. J Frail Aging 2016; 5: 204-7.
241.
Zahedi M, Salmani Izadi H, Arghidash F, Gumpricht E, Banach M, Sahebkar A. The effect of curcumin on hypoxia in the tumour microenvironment as a regulatory factor in cancer. Arch Med Sci 2023; 19: 1616-29.
242.
Banach M, Surma S. A look to the past – what has had the biggest impact on lipids in the last four decades? A personal perspective. Arch Med Sci 2023; 19: 559-64.
243.
Bielecka-Dabrowa A, Banach M, Wittczak A, et al. The role of nutraceuticals in heart failure muscle wasting as a result of inflammatory activity. The International Lipid Expert Panel (ILEP) Position Paper. Arch Med Sci 2023; 19: 841-64.
244.
Vukašinović A, Ostanek B, Klisic A, et al. Telomere-telomerase system status in patients with acute myocardial infarction with ST-segment elevation – relationship with oxidative stress. Arch Med Sci 2021; 19: 313-23.
245.
Ma L, Gao J. Suppression of lncRNA-MALAT1 activity ameliorates femoral head necrosis by modulating mTOR signaling. Arch Med Sci 2024; 20: 612-7.