Next Article in Journal
The Role of Selected Interleukins in the Development and Progression of Multiple Sclerosis—A Systematic Review
Next Article in Special Issue
Maternal Diet High in Linoleic Acid Alters Renal Branching Morphogenesis and mTOR/AKT Signalling Genes in Rat Fetal Kidneys
Previous Article in Journal
Changes in the Concentrations of Proangiogenic Cytokines in Human Brain Glioma and Acute Lymphoblastic Leukemia
Previous Article in Special Issue
The Role of mTOR in B Cell Lymphoid Malignancies: Biologic and Therapeutic Aspects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

mTOR Signaling: Recent Progress

by
Antonios N. Gargalionis
1,*,
Kostas A. Papavassiliou
2 and
Athanasios G. Papavassiliou
3,*
1
Department of Biopathology, ‘Eginition’ Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
2
‘Sotiria’ Hospital, Medical School, First University Department of Respiratory Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
3
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2587; https://doi.org/10.3390/ijms25052587
Submission received: 9 February 2024 / Accepted: 21 February 2024 / Published: 23 February 2024
(This article belongs to the Special Issue mTOR Signaling: Recent Progress)
In the intricate landscape of human biology, the mechanistic target of rapamycin (mTOR) emerges as a key regulator, orchestrating a vast array of processes in health and disease. mTOR is an evolutionarily conserved serine/threonine kinase that participates in multielement complexes with varying functional roles, depending on the established intermolecular associations and upstream cues [1]. The mTORC1 complex contains Raptor as a major component and responds to environmental/cellular stresses such as DNA damage and reactive oxygen species (ROS), the degree of the abundance of amino acids and growth factors, and energy from glucose and other sources [2,3]. mTORC1 mediates translation and protein synthesis, autophagy, lysosomal and mitochondrial biogenesis, lipid and nucleotide synthesis, mainly through upstream receptor tyrosine kinase (RTK) signaling. RTK concomitant kinase cascades include cytokine, insulin-like growth factor-1 (IGF-1), Wnt, phosphoinositide 3-kinase (PI3K)/AKT, mitogen-activated protein kinase (MAPK), and 5′ adenosine monophosphate-activated protein kinase (AMPK) pathways, most of which integrate into the tuberous sclerosis complex 1/2 (TSC1/TSC2) [4,5,6]. The mTORC2 complex is characterized by the presence of the Rictor component and regulates ion transport, cell survival, proliferation, migration, cytoskeleton remodeling, and glucose metabolism, mainly through IGF and PI3K/AKT signaling [7]. Furthermore, mTORC1 and mTORC2 are mutually regulated through diverse mTOR-associated signaling components, thus forming an intricate regulatory network that governs cellular homeostasis and disease pathogenesis [8].
The mechanisms though which mTOR specifically regulates autophagy, a cell survival process of cellular recycling, have been extensively highlighted at the intersection of cellular homeostasis for the cell to maintain intracellular balance and respond to environmental stresses. mTOR lies in the center of a complex interplay, where autophagy functions both upstream and downstream. Mechanistically, mTOR wields a dual role both by suppressing autophagy when activated by the abundance of cellular energetics, and also favoring the same process when suppressed, thereby enhancing the mechanisms that preserve cellular nutritional status [9]. This link becomes particularly interesting in cancer progression and treatment responses, since it is not clear yet whether activation or repression of autophagy is the established tumorigenic mechanism [10,11]. In certain types of malignancy, where autophagy promotes tumor progression, stemness, and drug resistance, pharmacological inhibition of autophagy is a therapeutic approach under investigation. Additionally, targeting mTOR-mediated autophagy has been proven to alleviate drug resistance, whereas drug resistance induced by mTOR suppresses autophagy and creates a favorable environment for the therapeutic exploitation of cancer metabolism [12,13].
Dysregulation of the mTOR pathway in cancer occurs due to genetic and epigenetic alterations, disturbed homeostasis of upstream regulators, and post-translational modifications [14]. During tumorigenesis, upregulation of oncogenes and downregulation of tumor-suppressors lead to hyperactivation of the mTOR signaling network in almost 30% of the neoplasms [1,15]. These alterations include mutations in the mTOR gene, in the Rheb and TSC1/TSC2 genes, amplification of Rictor and genetic defects of the upstream phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), KRAS, AKT, insulin-like growth factor receptor (IGFR), and epidermal growth factor receptor (EGFR) genes [1]. Moreover, mTOR signaling is implicated in functional traits of immune cells and immune signaling from dendritic cells to T cells, thereby modulating the tumor immune microenvironment [16,17]. mTOR pathway signaling components also respond to the tumor microenvironment (TME) mechanical stresses through mechanosensitive protein molecules and facilitate traits of cancer cells [2,18]. Several members of the pathway are regulated by long noncoding RNAs (lncRNAs) and also contribute decisively to self-renewal, tumor promotion, and drug resistance [1].
mTOR is also critically involved in regulating both normal physiological processes and aberrant functions within the central nervous system. The mTOR signaling axis controls the survival and development of brain cells and processes of learning and memory, as well as neuronal and synaptic plasticity [19]. mTOR is mechanistically tightly interconnected with the TSC1/TSC2 complex; therefore, it has been found upregulated in patients with TSC—an autosomal dominant disorder caused by loss-of-function mutations of either TSC1 or TSC2 genes—who develop neurological manifestations, including epilepsy, neuropsychiatric disorders, autism, and brain tumors [20,21]. mTOR inhibition in TSC patients is promising against epilepsy, whereas mTORC1-associated autophagy has been correlated with neurodegenerative disorders such as Alzheimer’s disease and Parkinson’s disease [22,23,24]. mTOR signaling is also engaged in the regulation of cardiac physiology and corresponding cardiovascular maladies. These include pulmonary arterial hypertension, myocardial infarction, and atherosclerosis [14].
Several small-molecule compounds have been discovered that target mTOR-triggered pathobiologies, especially in various types of malignancy [1]. These compounds have already taken their place in the clinic, including rapalogs (rapamycin derivatives) or mTOR inhibitors, such as Nab-sirolimus against metastatic or unresectable PEComas (tumors showing perivascular epithelioid cell differentiation) and lymphangioleiomyomatosis (LAM) [25,26,27], temsirolimus against advanced renal cell carcinoma (RCC) [28], everolimus for advanced RCC, advanced breast carcinoma and neuroendocrine pancreatic, lung, and gastrointestinal tract carcinomas [29,30,31,32], and ridaforolimus, which demonstrates efficacy against bone sarcomas [33]. Due to resistance to rapalogs and mTOR inhibitors, dual PI3K and mTOR inhibitors have been developed [34]. The mTOR pathway can also be targeted using ATP-competitive mTOR inhibitors, PI3K and AKT inhibitors [1].
Investigation of mTOR in human pathobiology has provided a wide spectrum of therapeutic opportunities, but also continuous controversies. mTOR exerts a multifaceted role in physiology and associated disorders, evoking debates about efficient mTOR pharmacological targeting. Deciphering these complex mechanisms of mTOR function and anomalous activity will provide new tools to fully utilize the dynamics of mTOR inhibition.

Author Contributions

Conceptualization, A.N.G. and A.G.P.; writing—original draft preparation, A.N.G. and K.A.P.; supervision, A.G.P.; writing—review and editing, A.G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Panwar, V.; Singh, A.; Bhatt, M.; Tonk, R.K.; Azizov, S.; Raza, A.S.; Sengupta, S.; Kumar, D.; Garg, M. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct. Target. Ther. 2023, 8, 375. [Google Scholar] [CrossRef]
  2. Gargalionis, A.N.; Papavassiliou, K.A.; Basdra, E.K.; Papavassiliou, A.G. mTOR Signaling Components in Tumor Mechanobiology. Int. J. Mol. Sci. 2022, 23, 1825. [Google Scholar] [CrossRef] [PubMed]
  3. Hara, K.; Maruki, Y.; Long, X.; Yoshino, K.; Oshiro, N.; Hidayat, S.; Tokunaga, C.; Avruch, J.; Yonezawa, K. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 2002, 110, 177–189. [Google Scholar] [CrossRef] [PubMed]
  4. Dai, X.; Jiang, C.; Jiang, Q.; Fang, L.; Yu, H.; Guo, J.; Yan, P.; Chi, F.; Zhang, T.; Inuzuka, H.; et al. AMPK-dependent phosphorylation of the GATOR2 component WDR24 suppresses glucose-mediated mTORC1 activation. Nat. Metab. 2023, 5, 265–276. [Google Scholar] [CrossRef] [PubMed]
  5. Menon, S.; Dibble, C.C.; Talbott, G.; Hoxhaj, G.; Valvezan, A.J.; Takahashi, H.; Cantley, L.C.; Manning, B.D. Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome. Cell 2014, 156, 771–785. [Google Scholar] [CrossRef]
  6. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [PubMed]
  7. Fu, W.; Hall, M.N. Regulation of mTORC2 Signaling. Genes 2020, 11, 1045. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, F.; Meng, M.; Mo, B.; Yang, Y.; Ji, Y.; Huang, P.; Lai, W.; Pan, X.; You, T.; Luo, H.; et al. Crosstalks between mTORC1 and mTORC2 variagate cytokine signaling to control NK maturation and effector function. Nat. Commun. 2018, 9, 4874. [Google Scholar] [CrossRef]
  9. Deleyto-Seldas, N.; Efeyan, A. The mTOR-Autophagy Axis and the Control of Metabolism. Front. Cell Dev. Biol. 2021, 9, 655731. [Google Scholar] [CrossRef]
  10. Liu, M.; Jiang, L.; Fu, X.; Wang, W.; Ma, J.; Tian, T.; Nan, K.; Liang, X. Cytoplasmic liver kinase B1 promotes the growth of human lung adenocarcinoma by enhancing autophagy. Cancer Sci. 2018, 109, 3055–3067. [Google Scholar] [CrossRef]
  11. Luo, T.; Fu, J.; Xu, A.; Su, B.; Ren, Y.; Li, N.; Zhu, J.; Zhao, X.; Dai, R.; Cao, J.; et al. PSMD10/gankyrin induces autophagy to promote tumor progression through cytoplasmic interaction with ATG7 and nuclear transactivation of ATG7 expression. Autophagy 2016, 12, 1355–1371. [Google Scholar] [CrossRef]
  12. Gremke, N.; Polo, P.; Dort, A.; Schneikert, J.; Elmshauser, S.; Brehm, C.; Klingmuller, U.; Schmitt, A.; Reinhardt, H.C.; Timofeev, O.; et al. mTOR-mediated cancer drug resistance suppresses autophagy and generates a druggable metabolic vulnerability. Nat. Commun. 2020, 11, 4684. [Google Scholar] [CrossRef]
  13. Xu, Z.; Han, X.; Ou, D.; Liu, T.; Li, Z.; Jiang, G.; Liu, J.; Zhang, J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl. Microbiol. Biotechnol. 2020, 104, 575–587. [Google Scholar] [CrossRef]
  14. Yu, J.J.; Goncharova, E.A. mTOR Signaling Network in Cell Biology and Human Disease. Int. J. Mol. Sci. 2022, 23, 16142. [Google Scholar] [CrossRef]
  15. Grabiner, B.C.; Nardi, V.; Birsoy, K.; Possemato, R.; Shen, K.; Sinha, S.; Jordan, A.; Beck, A.H.; Sabatini, D.M. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov. 2014, 4, 554–563. [Google Scholar] [CrossRef] [PubMed]
  16. Chi, H. Regulation and function of mTOR signalling in T cell fate decisions. Nat. Rev. Immunol. 2012, 12, 325–338. [Google Scholar] [CrossRef]
  17. Mafi, S.; Mansoori, B.; Taeb, S.; Sadeghi, H.; Abbasi, R.; Cho, W.C.; Rostamzadeh, D. mTOR-Mediated Regulation of Immune Responses in Cancer and Tumor Microenvironment. Front. Immunol. 2021, 12, 774103. [Google Scholar] [CrossRef] [PubMed]
  18. Gargalionis, A.N.; Sarlani, E.; Stofas, A.; Malakou, L.S.; Adamopoulos, C.; Bamias, A.; Boutati, E.; Constantinides, C.A.; Stravodimos, K.G.; Piperi, C.; et al. Polycystin-1 induces activation of the PI3K/AKT/mTOR pathway and promotes angiogenesis in renal cell carcinoma. Cancer Lett. 2020, 489, 135–143. [Google Scholar] [CrossRef] [PubMed]
  19. Lipton, J.O.; Sahin, M. The neurology of mTOR. Neuron 2014, 84, 275–291. [Google Scholar] [CrossRef]
  20. Tang, X.; Angst, G.; Haas, M.; Yang, F.; Wang, C. The Characterization of a Subependymal Giant Astrocytoma-Like Cell Line from Murine Astrocyte with mTORC1 Hyperactivation. Int. J. Mol. Sci. 2021, 22, 4116. [Google Scholar] [CrossRef]
  21. Zimmer, T.S.; Broekaart, D.W.M.; Gruber, V.E.; van Vliet, E.A.; Muhlebner, A.; Aronica, E. Tuberous Sclerosis Complex as Disease Model for Investigating mTOR-Related Gliopathy During Epileptogenesis. Front. Neurol. 2020, 11, 1028. [Google Scholar] [CrossRef]
  22. Bellozi, P.M.Q.; Gomes, G.F.; de Oliveira, L.R.; Olmo, I.G.; Vieira, E.L.M.; Ribeiro, F.M.; Fiebich, B.L.; de Oliveira, A.C.P. NVP-BEZ235 (Dactolisib) Has Protective Effects in a Transgenic Mouse Model of Alzheimer’s Disease. Front. Pharmacol. 2019, 10, 1345. [Google Scholar] [CrossRef]
  23. Smialek, D.; Kotulska, K.; Duda, A.; Jozwiak, S. Effect of mTOR Inhibitors in Epilepsy Treatment in Children with Tuberous Sclerosis Complex Under 2 Years of Age. Neurol. Ther. 2023, 12, 931–946. [Google Scholar] [CrossRef]
  24. Spilman, P.; Podlutskaya, N.; Hart, M.J.; Debnath, J.; Gorostiza, O.; Bredesen, D.; Richardson, A.; Strong, R.; Galvan, V. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS ONE 2010, 5, e9979. [Google Scholar] [CrossRef]
  25. Bissler, J.J.; McCormack, F.X.; Young, L.R.; Elwing, J.M.; Chuck, G.; Leonard, J.M.; Schmithorst, V.J.; Laor, T.; Brody, A.S.; Bean, J.; et al. Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N. Engl. J. Med. 2008, 358, 140–151. [Google Scholar] [CrossRef]
  26. McCormack, F.X.; Inoue, Y.; Moss, J.; Singer, L.G.; Strange, C.; Nakata, K.; Barker, A.F.; Chapman, J.T.; Brantly, M.L.; Stocks, J.M.; et al. Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N. Engl. J. Med. 2011, 364, 1595–1606. [Google Scholar] [CrossRef] [PubMed]
  27. Wagner, A.J.; Ravi, V.; Riedel, R.F.; Ganjoo, K.; Van Tine, B.A.; Chugh, R.; Cranmer, L.; Gordon, E.M.; Hornick, J.L.; Du, H.; et al. nab-Sirolimus for Patients With Malignant Perivascular Epithelioid Cell Tumors. J. Clin. Oncol. 2021, 39, 3660–3670. [Google Scholar] [CrossRef] [PubMed]
  28. Zanardi, E.; Verzoni, E.; Grassi, P.; Necchi, A.; Giannatempo, P.; Raggi, D.; De Braud, F.; Procopio, G. Clinical experience with temsirolimus in the treatment of advanced renal cell carcinoma. Ther. Adv. Urol. 2015, 7, 152–161. [Google Scholar] [CrossRef] [PubMed]
  29. Lee, L.; Ito, T.; Jensen, R.T. Everolimus in the treatment of neuroendocrine tumors: Efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence. Expert. Opin. Pharmacother. 2018, 19, 909–928. [Google Scholar] [CrossRef]
  30. O’Shaughnessy, J.; Thaddeus Beck, J.; Royce, M. Everolimus-based combination therapies for HR+, HER2- metastatic breast cancer. Cancer Treat. Rev. 2018, 69, 204–214. [Google Scholar] [CrossRef] [PubMed]
  31. Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M.; et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): A randomised, placebo-controlled, phase 3 study. Lancet 2016, 387, 968–977. [Google Scholar] [CrossRef] [PubMed]
  32. Yao, J.C.; Shah, M.H.; Ito, T.; Bohas, C.L.; Wolin, E.M.; Van Cutsem, E.; Hobday, T.J.; Okusaka, T.; Capdevila, J.; de Vries, E.G.; et al. Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 514–523. [Google Scholar] [CrossRef] [PubMed]
  33. Chawla, S.P.; Staddon, A.P.; Baker, L.H.; Schuetze, S.M.; Tolcher, A.W.; D’Amato, G.Z.; Blay, J.Y.; Mita, M.M.; Sankhala, K.K.; Berk, L.; et al. Phase II study of the mammalian target of rapamycin inhibitor ridaforolimus in patients with advanced bone and soft tissue sarcomas. J. Clin. Oncol. 2012, 30, 78–84. [Google Scholar] [CrossRef]
  34. Wu, X.; Xu, Y.; Liang, Q.; Yang, X.; Huang, J.; Wang, J.; Zhang, H.; Shi, J. Recent Advances in Dual PI3K/mTOR Inhibitors for Tumour Treatment. Front. Pharmacol. 2022, 13, 875372. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gargalionis, A.N.; Papavassiliou, K.A.; Papavassiliou, A.G. mTOR Signaling: Recent Progress. Int. J. Mol. Sci. 2024, 25, 2587. https://doi.org/10.3390/ijms25052587

AMA Style

Gargalionis AN, Papavassiliou KA, Papavassiliou AG. mTOR Signaling: Recent Progress. International Journal of Molecular Sciences. 2024; 25(5):2587. https://doi.org/10.3390/ijms25052587

Chicago/Turabian Style

Gargalionis, Antonios N., Kostas A. Papavassiliou, and Athanasios G. Papavassiliou. 2024. "mTOR Signaling: Recent Progress" International Journal of Molecular Sciences 25, no. 5: 2587. https://doi.org/10.3390/ijms25052587

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

Article Metrics

Back to TopTop