Cell and Cell-Free Therapies to Counteract Human Premature and Physiological Aging: MSCs Come to Light
Abstract
:1. Introduction
2. Progeroid Laminopathies: A Model to Study the Relevance of MSCs in Aging
3. MSCs as a Therapy for Aging
3.1. MSCs Therapies to Counteract Physiological Aging
3.1.1. Knee Osteoarthritis
3.1.2. Cardiovascular Disease
3.1.3. Aging Frailty
3.2. MSCs to Counteract Premature Aging Syndromes
4. MSC-Based Cell-Free Therapy for Aging
5. Harnessing the Efficacy of MSC-Based Therapies
6. Future Directions and Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
- Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef] [Green Version]
- Prince, M.J.; Wu, F.; Guo, Y.; Gutierrez Robledo, L.M.; O′Donnell, M.; Sullivan, R.; Yusuf, S. The burden of disease in older people and implications for health policy and practice. Lancet 2015, 385, 549–562. [Google Scholar] [CrossRef]
- Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Ravikumar, M.; Ling, L.; Nurcombe, V.; Cool, S.M. Age-Related Changes in the Inflammatory Status of Human Mesenchymal Stem Cells: Implications for Cell Therapy. Stem Cell Rep. 2021, 16, 694–707. [Google Scholar] [CrossRef] [PubMed]
- Baxter, M.A.; Wynn, R.F.; Jowitt, S.N.; Wraith, J.E.; Fairbairn, L.J.; Bellantuono, I. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 2004, 22, 675–682. [Google Scholar] [CrossRef]
- Choudhery, M.S.; Badowski, M.; Muise, A.; Pierce, J.; Harris, D.T. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J. Transl. Med. 2014, 12, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.Y.; Yu, K.R.; Lee, B.C.; Kang, I.; Kim, J.J.; Jung, E.J.; Kim, H.S.; Seo, Y.; Choi, S.W.; Kang, K.S. GATA4-dependent regulation of the secretory phenotype via MCP-1 underlies lamin A-mediated human mesenchymal stem cell aging. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Levy, O.; Kuai, R.; Siren, E.M.J.; Bhere, D.; Milton, Y.; Nissar, N.; De Biasio, M.; Heinelt, M.; Reeve, B.; Abdi, R.; et al. Shattering barriers toward clinically meaningful MSC therapies. Sci. Adv. 2020, 6, eaba6884. [Google Scholar] [CrossRef]
- Pittenger, M.F.; Discher, D.E.; Péault, B.M.; Phinney, D.G.; Hare, J.M.; Caplan, A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen. Med. 2019, 4, 22. [Google Scholar] [CrossRef] [Green Version]
- Horwitz, E.M.; Gordon, P.L.; Koo, W.K.; Marx, J.C.; Neel, M.D.; McNall, R.Y.; Muul, L.; Hofmann, T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc. Natl. Acad. Sci. USA 2002, 99, 8932–8937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Otsuru, S.; Desbourdes, L.; Guess, A.J.; Hofmann, T.J.; Relation, T.; Kaito, T.; Dominici, M.; Iwamoto, M.; Horwitz, E.M. Extracellular vesicles released from mesenchymal stromal cells stimulate bone growth in osteogenesis imperfecta. Cytotherapy 2018, 20, 62–73. [Google Scholar] [CrossRef] [PubMed]
- Infante, A.; Gener, B.; Vázquez, M.; Olivares, N.; Arrieta, A.; Grau, G.; Llano, I.; Madero, L.; Bueno, A.M.; Sagastizabal, B.; et al. Reiterative infusions of MSCs improve pediatric osteogenesis imperfecta eliciting a pro-osteogenic paracrine response: TERCELOI clinical trial. Clin. Transl. Med. 2021, 11, e265. [Google Scholar] [CrossRef]
- Moghadasi, S.; Elveny, M.; Rahman, H.S.; Suksatan, W.; Jalil, A.T.; Abdelbasset, W.K.; Yumashev, A.V.; Shariatzadeh, S.; Motavalli, R.; Behzad, F.; et al. A paradigm shift in cell-free approach: The emerging role of MSCs-derived exosomes in regenerative medicine. J. Transl. Med. 2021, 19, 302. [Google Scholar] [CrossRef] [PubMed]
- Oeppen, J.; Vaupel, J.W. Demography. Broken limits to life expectancy. Science 2002, 296, 1029–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scott, A.J.; Ellison, M.; Sinclair, D.A. The economic value of targeting aging. Nat. Aging 2021, 1, 616–623. [Google Scholar] [CrossRef]
- Carrero, D.; Soria-Valles, C.; López-Otín, C. Hallmarks of progeroid syndromes: Lessons from mice and reprogrammed cells. Dis. Model. Mech. 2016, 9, 719–735. [Google Scholar] [CrossRef] [Green Version]
- Goldman, R.D.; Gruenbaum, Y.; Moir, R.D.; Shumaker, D.K.; Spann, T.P. Nuclear lamins: Building blocks of nuclear architecture. Genes Dev. 2002, 16, 533–547. [Google Scholar] [CrossRef] [Green Version]
- Marcelot, A.; Worman, H.J.; Zinn-Justin, S. Protein structural and mechanistic basis of progeroid laminopathies. FEBS J. 2021, 288, 2757–2772. [Google Scholar] [CrossRef]
- Merideth, M.A.; Gordon, L.B.; Clauss, S.; Sachdev, V.; Smith, A.C.; Perry, M.B.; Brewer, C.C.; Zalewski, C.; Kim, H.J.; Solomon, B.; et al. Phenotype and course of Hutchinson-Gilford progeria syndrome. N. Engl. J. Med. 2008, 358, 592–604. [Google Scholar] [CrossRef] [Green Version]
- Gordon, L.B.; Shappell, H.; Massaro, J.; D′Agostino, R.B.; Brazier, J.; Campbell, S.E.; Kleinman, M.E.; Kieran, M.W. Association of Lonafarnib Treatment vs No Treatment With Mortality Rate in Patients With Hutchinson-Gilford Progeria Syndrome. JAMA 2018, 319, 1687–1695. [Google Scholar] [CrossRef] [Green Version]
- Bergo, M.O.; Gavino, B.; Ross, J.; Schmidt, W.K.; Hong, C.; Kendall, L.V.; Mohr, A.; Meta, M.; Genant, H.; Jiang, Y.; et al. Zmpste24 deficiency in mice causes spontaneous bone fractures, muscle weakness, and a prelamin A processing defect. Proc. Natl. Acad. Sci. USA 2002, 99, 13049–13054. [Google Scholar] [CrossRef] [Green Version]
- Osorio, F.G.; Navarro, C.L.; Cadiñanos, J.; López-Mejía, I.C.; Quirós, P.M.; Bartoli, C.; Rivera, J.; Tazi, J.; Guzmán, G.; Varela, I.; et al. Splicing-directed therapy in a new mouse model of human accelerated aging. Sci. Transl. Med. 2011, 3, 106ra107. [Google Scholar] [CrossRef]
- Infante, A.; Rodríguez, C.I. Osteogenesis and aging: Lessons from mesenchymal stem cells. Stem Cell Res. Ther. 2018, 9, 244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scaffidi, P.; Misteli, T. Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat. Cell Biol. 2008, 10, 452–459. [Google Scholar] [CrossRef] [Green Version]
- Ruiz de Eguino, G.; Infante, A.; Schlangen, K.; Aransay, A.M.; Fullaondo, A.; Soriano, M.; García-Verdugo, J.M.; Martín, A.G.; Rodríguez, C.I. Sp1 transcription factor interaction with accumulated prelamin a impairs adipose lineage differentiation in human mesenchymal stem cells: Essential role of sp1 in the integrity of lipid vesicles. Stem Cells Transl. Med. 2012, 1, 309–321. [Google Scholar] [CrossRef]
- Infante, A.; Gago, A.; de Eguino, G.R.; Calvo-Fernández, T.; Gómez-Vallejo, V.; Llop, J.; Schlangen, K.; Fullaondo, A.; Aransay, A.M.; Martín, A.; et al. Prelamin A accumulation and stress conditions induce impaired Oct-1 activity and autophagy in prematurely aged human mesenchymal stem cell. Aging 2014, 6, 264–280. [Google Scholar] [CrossRef] [Green Version]
- Infante, A.; Rodríguez, C.I. Secretome analysis of in vitro aged human mesenchymal stem cells reveals IGFBP7 as a putative factor for promoting osteogenesis. Sci. Rep. 2018, 8, 4632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Lian, Q.; Zhu, G.; Zhou, F.; Sui, L.; Tan, C.; Mutalif, R.A.; Navasankari, R.; Zhang, Y.; Tse, H.F.; et al. A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 2011, 8, 31–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sánchez, P.; Infante, A.; de Eguino, G.R.; Fuentes-Maestre, J.A.; García-Verdugo, J.M.; Rodríguez, C.I. Age-Related Lipid Metabolic Signature in Human LMNA-Lipodystrophic Stem Cell-Derived Adipocytes. J. Clin. Endocrinol. Metab. 2015, 100, E964–E973. [Google Scholar] [CrossRef] [Green Version]
- Xiong, Z.M.; LaDana, C.; Wu, D.; Cao, K. An inhibitory role of progerin in the gene induction network of adipocyte differentiation from iPS cells. Aging 2013, 5, 288–303. [Google Scholar] [CrossRef] [Green Version]
- Blondel, S.; Jaskowiak, A.L.; Egesipe, A.L.; Le Corf, A.; Navarro, C.; Cordette, V.; Martinat, C.; Laabi, Y.; Djabali, K.; de Sandre-Giovannoli, A.; et al. Induced pluripotent stem cells reveal functional differences between drugs currently investigated in patients with hutchinson-gilford progeria syndrome. Stem Cells Transl. Med. 2014, 3, 510–519. [Google Scholar] [CrossRef] [PubMed]
- Lo Cicero, A.; Jaskowiak, A.L.; Egesipe, A.L.; Tournois, J.; Brinon, B.; Pitrez, P.R.; Ferreira, L.; de Sandre-Giovannoli, A.; Levy, N.; Nissan, X. A High Throughput Phenotypic Screening reveals compounds that counteract premature osteogenic differentiation of HGPS iPS-derived mesenchymal stem cells. Sci. Rep. 2016, 6, 34798. [Google Scholar] [CrossRef]
- Choi, J.Y.; Lai, J.K.; Xiong, Z.M.; Ren, M.; Moorer, M.C.; Stains, J.P.; Cao, K. Diminished Canonical β-Catenin Signaling During Osteoblast Differentiation Contributes to Osteopenia in Progeria. J. Bone Min. Res. 2018, 33, 2059–2070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egesipe, A.L.; Blondel, S.; Lo Cicero, A.; Jaskowiak, A.L.; Navarro, C.; Sandre-Giovannoli, A.; Levy, N.; Peschanski, M.; Nissan, X. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson-Gilford progeria syndrome cells. NPJ Aging Mech. Dis. 2016, 2, 16026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scaffidi, P.; Misteli, T. Lamin A-dependent nuclear defects in human aging. Science 2006, 312, 1059–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ragnauth, C.D.; Warren, D.T.; Liu, Y.; McNair, R.; Tajsic, T.; Figg, N.; Shroff, R.; Skepper, J.; Shanahan, C.M. Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation 2010, 121, 2200–2210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, K.; Blair, C.D.; Faddah, D.A.; Kieckhaefer, J.E.; Olive, M.; Erdos, M.R.; Nabel, E.G.; Collins, F.S. Progerin and telomere dysfunction collaborate to trigger cellular senescence in normal human fibroblasts. J. Clin. Invest. 2011, 121, 2833–2844. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Drozdov, I.; Shroff, R.; Beltran, L.E.; Shanahan, C.M. Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells. Circ. Res. 2013, 112, e99–e109. [Google Scholar] [CrossRef] [Green Version]
- Qu, Y.N.; Zhang, L.; Wang, T.; Zhang, H.Y.; Yang, Z.J.; Yuan, F.F.; Wang, Y.; Li, S.W.; Jiang, X.X.; Xie, X.H. Vitamin C Treatment Rescues Prelamin A-Induced Premature Senescence of Subchondral Bone Mesenchymal Stem Cells. Stem Cells Int. 2020, 2020, 3150716. [Google Scholar] [CrossRef]
- Farr, J.N.; Xu, M.; Weivoda, M.M.; Monroe, D.G.; Fraser, D.G.; Onken, J.L.; Negley, B.A.; Sfeir, J.G.; Ogrodnik, M.B.; Hachfeld, C.M.; et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat. Med. 2017, 23, 1072–1079. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Palmer, A.K.; Ding, H.; Weivoda, M.M.; Pirtskhalava, T.; White, T.A.; Sepe, A.; Johnson, K.O.; Stout, M.B.; Giorgadze, N.; et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife 2015, 4, e12997. [Google Scholar] [CrossRef] [PubMed]
- Martin-Montalvo, A.; Mercken, E.M.; Mitchell, S.J.; Palacios, H.H.; Mote, P.L.; Scheibye-Knudsen, M.; Gomes, A.P.; Ward, T.M.; Minor, R.K.; Blouin, M.J.; et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 2013, 4, 2192. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Xin, X.; Wang, L.; Wang, B.; Chen, L.; Liu, O.; Rowe, D.W.; Xu, M. Senolytics improve bone forming potential of bone marrow mesenchymal stem cells from aged mice. NPJ Regen. Med. 2021, 6, 34. [Google Scholar] [CrossRef] [PubMed]
- Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [Green Version]
- Conboy, I.M.; Conboy, M.J.; Wagers, A.J.; Girma, E.R.; Weissman, I.L.; Rando, T.A. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005, 433, 760–764. [Google Scholar] [CrossRef]
- Villeda, S.A.; Plambeck, K.E.; Middeldorp, J.; Castellano, J.M.; Mosher, K.I.; Luo, J.; Smith, L.K.; Bieri, G.; Lin, K.; Berdnik, D.; et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 2014, 20, 659–663. [Google Scholar] [CrossRef] [Green Version]
- Castellano, J.M.; Mosher, K.I.; Abbey, R.J.; McBride, A.A.; James, M.L.; Berdnik, D.; Shen, J.C.; Zou, B.; Xie, X.S.; Tingle, M.; et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 2017, 544, 488–492. [Google Scholar] [CrossRef] [PubMed]
- Villeda, S.A.; Luo, J.; Mosher, K.I.; Zou, B.; Britschgi, M.; Bieri, G.; Stan, T.M.; Fainberg, N.; Ding, Z.; Eggel, A.; et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011, 477, 90–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horowitz, A.M.; Fan, X.; Bieri, G.; Smith, L.K.; Sanchez-Diaz, C.I.; Schroer, A.B.; Gontier, G.; Casaletto, K.B.; Kramer, J.H.; Williams, K.E.; et al. Blood factors transfer beneficial effects of exercise on neurogenesis and cognition to the aged brain. Science 2020, 369, 167–173. [Google Scholar] [CrossRef] [PubMed]
- Josephson, A.M.; Bradaschia-Correa, V.; Lee, S.; Leclerc, K.; Patel, K.S.; Muinos Lopez, E.; Litwa, H.P.; Neibart, S.S.; Kadiyala, M.; Wong, M.Z.; et al. Age-related inflammation triggers skeletal stem/progenitor cell dysfunction. Proc. Natl. Acad. Sci. USA 2019, 116, 6995–7004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef]
- Squarzoni, S.; Schena, E.; Sabatelli, P.; Mattioli, E.; Capanni, C.; Cenni, V.; D’Apice, M.R.; Andrenacci, D.; Sarli, G.; Pellegrino, V.; et al. Interleukin-6 neutralization ameliorates symptoms in prematurely aged mice. Aging Cell 2021, 20, e13285. [Google Scholar] [CrossRef]
- Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
- Pelletier, J.P.; Martel-Pelletier, J.; Abramson, S.B. Osteoarthritis, an inflammatory disease: Potential implication for the selection of new therapeutic targets. Arthritis Rheum. 2001, 44, 1237–1247. [Google Scholar] [CrossRef]
- O’Brien, K.; Tailor, P.; Leonard, C.; DiFrancesco, L.M.; Hart, D.A.; Matyas, J.R.; Frank, C.B.; Krawetz, R.J. Enumeration and Localization of Mesenchymal Progenitor Cells and Macrophages in Synovium from Normal Individuals and Patients with Pre-Osteoarthritis or Clinically Diagnosed Osteoarthritis. Int. J. Mol. Sci. 2017, 18, 774. [Google Scholar] [CrossRef] [Green Version]
- Vega, A.; Martín-Ferrero, M.A.; Del Canto, F.; Alberca, M.; García, V.; Munar, A.; Orozco, L.; Soler, R.; Fuertes, J.J.; Huguet, M.; et al. Treatment of Knee Osteoarthritis With Allogeneic Bone Marrow Mesenchymal Stem Cells: A Randomized Controlled Trial. Transplantation 2015, 99, 1681–1690. [Google Scholar] [CrossRef]
- Chahal, J.; Gómez-Aristizábal, A.; Shestopaloff, K.; Bhatt, S.; Chaboureau, A.; Fazio, A.; Chisholm, J.; Weston, A.; Chiovitti, J.; Keating, A.; et al. Bone Marrow Mesenchymal Stromal Cell Treatment in Patients with Osteoarthritis Results in Overall Improvement in Pain and Symptoms and Reduces Synovial Inflammation. Stem Cells Transl. Med. 2019, 8, 746–757. [Google Scholar] [CrossRef] [Green Version]
- Matas, J.; Orrego, M.; Amenabar, D.; Infante, C.; Tapia-Limonchi, R.; Cadiz, M.I.; Alcayaga-Miranda, F.; González, P.L.; Muse, E.; Khoury, M.; et al. Umbilical Cord-Derived Mesenchymal Stromal Cells (MSCs) for Knee Osteoarthritis: Repeated MSC Dosing Is Superior to a Single MSC Dose and to Hyaluronic Acid in a Controlled Randomized Phase I/II Trial. Stem Cells Transl. Med. 2019, 8, 215–224. [Google Scholar] [CrossRef] [Green Version]
- Jo, C.H.; Lee, Y.G.; Shin, W.H.; Kim, H.; Chai, J.W.; Jeong, E.C.; Kim, J.E.; Shim, H.; Shin, J.S.; Shin, I.S.; et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: A proof-of-concept clinical trial. Stem Cells 2014, 32, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
- Gupta, P.K.; Chullikana, A.; Rengasamy, M.; Shetty, N.; Pandey, V.; Agarwal, V.; Wagh, S.Y.; Vellotare, P.K.; Damodaran, D.; Viswanathan, P.; et al. Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®): Preclinical and clinical trial in osteoarthritis of the knee joint. Arthritis Res. Ther. 2016, 18, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamo-Espinosa, J.M.; Mora, G.; Blanco, J.F.; Granero-Moltó, F.; Nuñez-Córdoba, J.M.; Sánchez-Echenique, C.; Bondía, J.M.; Aquerreta, J.D.; Andreu, E.J.; Ornilla, E.; et al. Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: Multicenter randomized controlled clinical trial (phase I/II). J. Transl. Med. 2016, 14, 246. [Google Scholar] [CrossRef] [Green Version]
- Townsend, N.; Wilson, L.; Bhatnagar, P.; Wickramasinghe, K.; Rayner, M.; Nichols, M. Cardiovascular disease in Europe: Epidemiological update 2016. Eur. Heart J. 2016, 37, 3232–3245. [Google Scholar] [CrossRef]
- Pei, Z.; Zeng, J.; Song, Y.; Gao, Y.; Wu, R.; Chen, Y.; Li, F.; Li, W.; Zhou, H.; Yang, Y. In vivo imaging to monitor differentiation and therapeutic effects of transplanted mesenchymal stem cells in myocardial infarction. Sci. Rep. 2017, 7, 6296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hatzistergos, K.E.; Saur, D.; Seidler, B.; Balkan, W.; Breton, M.; Valasaki, K.; Takeuchi, L.M.; Landin, A.M.; Khan, A.; Hare, J.M. Stimulatory Effects of Mesenchymal Stem Cells on cKit+ Cardiac Stem Cells Are Mediated by SDF1/CXCR4 and SCF/cKit Signaling Pathways. Circ. Res. 2016, 119, 921–930. [Google Scholar] [CrossRef] [Green Version]
- Ohnishi, S.; Sumiyoshi, H.; Kitamura, S.; Nagaya, N. Mesenchymal stem cells attenuate cardiac fibroblast proliferation and collagen synthesis through paracrine actions. FEBS Lett. 2007, 581, 3961–3966. [Google Scholar] [CrossRef] [Green Version]
- Tompkins, B.A.; Balkan, W.; Winkler, J.; Gyöngyösi, M.; Goliasch, G.; Fernández-Avilés, F.; Hare, J.M. Preclinical Studies of Stem Cell Therapy for Heart Disease. Circ. Res. 2018, 122, 1006–1020. [Google Scholar] [CrossRef]
- Perin, E.C.; Borow, K.M.; Silva, G.V.; DeMaria, A.N.; Marroquin, O.C.; Huang, P.P.; Traverse, J.H.; Krum, H.; Skerrett, D.; Zheng, Y.; et al. A Phase II Dose-Escalation Study of Allogeneic Mesenchymal Precursor Cells in Patients With Ischemic or Nonischemic Heart Failure. Circ. Res. 2015, 117, 576–584. [Google Scholar] [CrossRef] [Green Version]
- Heldman, A.W.; DiFede, D.L.; Fishman, J.E.; Zambrano, J.P.; Trachtenberg, B.H.; Karantalis, V.; Mushtaq, M.; Williams, A.R.; Suncion, V.Y.; McNiece, I.K.; et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: The TAC-HFT randomized trial. JAMA 2014, 311, 62–73. [Google Scholar] [CrossRef]
- Bartolucci, J.; Verdugo, F.J.; González, P.L.; Larrea, R.E.; Abarzua, E.; Goset, C.; Rojo, P.; Palma, I.; Lamich, R.; Pedreros, P.A.; et al. Safety and Efficacy of the Intravenous Infusion of Umbilical Cord Mesenchymal Stem Cells in Patients With Heart Failure: A Phase 1/2 Randomized Controlled Trial (RIMECARD Trial [Randomized Clinical Trial of Intravenous Infusion Umbilical Cord Mesenchymal Stem Cells on Cardiopathy]). Circ. Res. 2017, 121, 1192–1204. [Google Scholar] [CrossRef]
- Bolli, R.; Mitrani, R.D.; Hare, J.M.; Pepine, C.J.; Perin, E.C.; Willerson, J.T.; Traverse, J.H.; Henry, T.D.; Yang, P.C.; Murphy, M.P.; et al. A Phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: The CCTRN CONCERT-HF trial. Eur. J. Heart Fail. 2021, 23, 661–674. [Google Scholar] [CrossRef]
- Hare, J.M.; DiFede, D.L.; Rieger, A.C.; Florea, V.; Landin, A.M.; El-Khorazaty, J.; Khan, A.; Mushtaq, M.; Lowery, M.H.; Byrnes, J.J.; et al. Randomized Comparison of Allogeneic Versus Autologous Mesenchymal Stem Cells for Nonischemic Dilated Cardiomyopathy: POSEIDON-DCM Trial. J. Am. Coll. Cardiol. 2017, 69, 526–537. [Google Scholar] [CrossRef]
- Florea, V.; Rieger, A.C.; DiFede, D.L.; El-Khorazaty, J.; Natsumeda, M.; Banerjee, M.N.; Tompkins, B.A.; Khan, A.; Schulman, I.H.; Landin, A.M.; et al. Dose Comparison Study of Allogeneic Mesenchymal Stem Cells in Patients With Ischemic Cardiomyopathy (The TRIDENT Study). Circ. Res. 2017, 121, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
- Ding, S.; Feng, L.; Wu, J.; Zhu, F.; Tan, Z.e.; Yao, R. Bioprinting of Stem Cells: Interplay of Bioprinting Process, Bioinks, and Stem Cell Properties. ACS Biomater. Sci. Eng. 2018, 4, 3108–3124. [Google Scholar] [CrossRef] [PubMed]
- Gordon, E.H.; Hubbard, R.E. Differences in frailty in older men and women. Med. J. Aust. 2020, 212, 183–188. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Ge, J.; Huang, C.; Liu, H.; Jiang, H. Application of mesenchymal stem cell therapy for aging frailty: From mechanisms to therapeutics. Theranostics 2021, 11, 5675–5685. [Google Scholar] [CrossRef]
- Golpanian, S.; DiFede, D.L.; Khan, A.; Schulman, I.H.; Landin, A.M.; Tompkins, B.A.; Heldman, A.W.; Miki, R.; Goldstein, B.J.; Mushtaq, M.; et al. Allogeneic Human Mesenchymal Stem Cell Infusions for Aging Frailty. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1505–1512. [Google Scholar] [CrossRef] [PubMed]
- Tompkins, B.A.; DiFede, D.L.; Khan, A.; Landin, A.M.; Schulman, I.H.; Pujol, M.V.; Heldman, A.W.; Miki, R.; Goldschmidt-Clermont, P.J.; Goldstein, B.J.; et al. Allogeneic Mesenchymal Stem Cells Ameliorate Aging Frailty: A Phase II Randomized, Double-Blind, Placebo-Controlled Clinical Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1513–1522. [Google Scholar] [CrossRef]
- Chinnadurai, R.; Ogedengbe, O.; Agarwal, P.; Money-Coomes, S.; Abdurrahman, A.Z.; Mohammed, S.; Kalra, P.A.; Rothwell, N.; Pradhan, S. Older age and frailty are the chief predictors of mortality in COVID-19 patients admitted to an acute medical unit in a secondary care setting- a cohort study. BMC Geriatr. 2020, 20, 409. [Google Scholar] [CrossRef]
- Bielza, R.; Sanz, J.; Zambrana, F.; Arias, E.; Malmierca, E.; Portillo, L.; Thuissard, I.J.; Lung, A.; Neira, M.; Moral, M.; et al. Clinical Characteristics, Frailty, and Mortality of Residents With COVID-19 in Nursing Homes of a Region of Madrid. J. Am. Med. Dir. Assoc. 2021, 22, 245–252.e242. [Google Scholar] [CrossRef]
- Zaki, M.M.; Lesha, E.; Said, K.; Kiaee, K.; Robinson-McCarthy, L.; George, H.; Hanna, A.; Appleton, E.; Liu, S.; Ng, A.H.M.; et al. Cell therapy strategies for COVID-19: Current approaches and potential applications. Sci. Adv. 2021, 7. [Google Scholar] [CrossRef]
- Wang, W.; Lei, W.; Jiang, L.; Gao, S.; Hu, S.; Zhao, Z.G.; Niu, C.Y.; Zhao, Z.A. Therapeutic mechanisms of mesenchymal stem cells in acute respiratory distress syndrome reveal potentials for Covid-19 treatment. J. Transl. Med. 2021, 19, 198. [Google Scholar] [CrossRef]
- Leng, Z.; Zhu, R.; Hou, W.; Feng, Y.; Yang, Y.; Han, Q.; Shan, G.; Meng, F.; Du, D.; Wang, S.; et al. Transplantation of ACE2. Aging Dis. 2020, 11, 216–228. [Google Scholar] [CrossRef] [Green Version]
- Schäfer, R.; Spohn, G.; Bechtel, M.; Bojkova, D.; Baer, P.C.; Kuçi, S.; Seifried, E.; Ciesek, S.; Cinatl, J. Human Mesenchymal Stromal Cells Are Resistant to SARS-CoV-2 Infection under Steady-State, Inflammatory Conditions and in the Presence of SARS-CoV-2-Infected Cells. Stem Cell Rep. 2021, 16, 419–427. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Guijo, F.; García-Arranz, M.; López-Parra, M.; Monedero, P.; Mata-Martínez, C.; Santos, A.; Sagredo, V.; Álvarez-Avello, J.M.; Guerrero, J.E.; Pérez-Calvo, C.; et al. Adipose-derived mesenchymal stromal cells for the treatment of patients with severe SARS-CoV-2 pneumonia requiring mechanical ventilation. A proof of concept study. EClinicalMedicine 2020, 25, 100454. [Google Scholar] [CrossRef]
- Lanzoni, G.; Linetsky, E.; Correa, D.; Messinger Cayetano, S.; Alvarez, R.A.; Kouroupis, D.; Alvarez Gil, A.; Poggioli, R.; Ruiz, P.; Marttos, A.C.; et al. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl. Med. 2021, 10, 660–673. [Google Scholar] [CrossRef] [PubMed]
- González-Dominguez, A.; Montañez, R.; Castejón-Vega, B.; Nuñez-Vasco, J.; Lendines-Cordero, D.; Wang, C.; Mbalaviele, G.; Navarro-Pando, J.M.; Alcocer-Gómez, E.; Cordero, M.D. Inhibition of the NLRP3 inflammasome improves lifespan in animal murine model of Hutchinson-Gilford Progeria. EMBO Mol. Med. 2021, 13, e14012. [Google Scholar] [CrossRef] [PubMed]
- Pak, J.; Lee, J.H.; Jeon, J.H.; Kim, Y.B.; Jeong, B.C.; Lee, S.H. Potential Benefits of Allogeneic Haploidentical Adipose Tissue-Derived Stromal Vascular Fraction in a Hutchinson-Gilford Progeria Syndrome Patient. Front. Bioeng. Biotechnol. 2020, 8, 574010. [Google Scholar] [CrossRef] [PubMed]
- Lundberg, E.; Kriström, B.; Jonsson, B.; Albertsson-Wikland, K.; group, s. Growth hormone (GH) dose-dependent IGF-I response relates to pubertal height gain. BMC Endocr. Disord. 2015, 15, 84. [Google Scholar] [CrossRef] [Green Version]
- Mariño, G.; Ugalde, A.P.; Fernández, A.F.; Osorio, F.G.; Fueyo, A.; Freije, J.M.; López-Otín, C. Insulin-like growth factor 1 treatment extends longevity in a mouse model of human premature aging by restoring somatotroph axis function. Proc. Natl. Acad. Sci. USA 2010, 107, 16268–16273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lei, Q.; Gao, F.; Liu, T.; Ren, W.; Chen, L.; Cao, Y.; Chen, W.; Guo, S.; Zhang, Q.; Wang, H.; et al. Extracellular vesicles deposit. Sci. Transl. Med. 2021, 13. [Google Scholar] [CrossRef]
- Grenier-Pleau, I.; Tyryshkin, K.; Le, T.D.; Rudan, J.; Bonneil, E.; Thibault, P.; Zeng, K.; Lässer, C.; Mallinson, D.; Lamprou, D.; et al. Blood extracellular vesicles from healthy individuals regulate hematopoietic stem cells as humans age. Aging Cell 2020, 19, e13245. [Google Scholar] [CrossRef] [PubMed]
- Fafián-Labora, J.A.; Rodríguez-Navarro, J.A.; O′Loghlen, A. Small Extracellular Vesicles Have GST Activity and Ameliorate Senescence-Related Tissue Damage. Cell Metab. 2020, 32, 71–86.e75. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Mahairaki, V.; Bai, H.; Ding, Z.; Li, J.; Witwer, K.W.; Cheng, L. Highly Purified Human Extracellular Vesicles Produced by Stem Cells Alleviate Aging Cellular Phenotypes of Senescent Human Cells. Stem Cells 2019, 37, 779–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, R.; Qin, C.; Wang, J.; Hu, Y.; Zheng, G.; Qiu, G.; Ge, M.; Tao, H.; Shu, Q.; Xu, J. Differential effects of extracellular vesicles from aging and young mesenchymal stem cells in acute lung injury. Aging 2019, 11, 7996–8014. [Google Scholar] [CrossRef] [PubMed]
- Dorronsoro, A.; Santiago, F.E.; Grassi, D.; Zhang, T.; Lai, R.C.; McGowan, S.J.; Angelini, L.; Lavasani, M.; Corbo, L.; Lu, A.; et al. Mesenchymal stem cell-derived extracellular vesicles reduce senescence and extend health span in mouse models of aging. Aging Cell 2021, 20, e13337. [Google Scholar] [CrossRef]
- Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef]
- Munir, S.; Basu, A.; Maity, P.; Krug, L.; Haas, P.; Jiang, D.; Strauss, G.; Wlaschek, M.; Geiger, H.; Singh, K.; et al. TLR4-dependent shaping of the wound site by MSCs accelerates wound healing. EMBO Rep. 2020, 21, e48777. [Google Scholar] [CrossRef]
- Peterson, K.M.; Aly, A.; Lerman, A.; Lerman, L.O.; Rodriguez-Porcel, M. Improved survival of mesenchymal stromal cell after hypoxia preconditioning: Role of oxidative stress. Life Sci. 2011, 88, 65–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, D.S.; Jang, I.K.; Lee, M.W.; Ko, Y.J.; Lee, D.H.; Lee, J.W.; Sung, K.W.; Koo, H.H.; Yoo, K.H. Enhanced Immunosuppressive Properties of Human Mesenchymal Stem Cells Primed by Interferon-γ. EBioMedicine 2018, 28, 261–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krampera, M.; Cosmi, L.; Angeli, R.; Pasini, A.; Liotta, F.; Andreini, A.; Santarlasci, V.; Mazzinghi, B.; Pizzolo, G.; Vinante, F.; et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006, 24, 386–398. [Google Scholar] [CrossRef]
- Chinnadurai, R.; Rajan, D.; Ng, S.; McCullough, K.; Arafat, D.; Waller, E.K.; Anderson, L.J.; Gibson, G.; Galipeau, J. Immune dysfunctionality of replicative senescent mesenchymal stromal cells is corrected by IFNγ priming. Blood Adv. 2017, 1, 628–643. [Google Scholar] [CrossRef] [PubMed]
- Kim, E.H.; Kim, D.H.; Kim, H.R.; Kim, S.Y.; Kim, H.H.; Bang, O.Y. Stroke Serum Priming Modulates Characteristics of Mesenchymal Stromal Cells by Controlling the Expression miRNA-20a. Cell Transplant. 2016, 25, 1489–1499. [Google Scholar] [CrossRef] [Green Version]
- Silva-Carvalho, A.; Rodrigues, L.P.; Schiavinato, J.L.; Alborghetti, M.R.; Bettarello, G.; Simões, B.P.; Neves, F.A.R.; Panepucci, R.A.; de Carvalho, J.L.; Saldanha-Araujo, F. GVHD-derived plasma as a priming strategy of mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 156. [Google Scholar] [CrossRef]
- Wang, R.; Wang, X.; Yang, S.; Xiao, Y.; Jia, Y.; Zhong, J.; Gao, Q.; Zhang, X. Umbilical cord-derived mesenchymal stem cells promote myeloid-derived suppressor cell enrichment by secreting CXCL1 to prevent graft-versus-host disease after hematopoietic stem cell transplantation. Cytotherapy 2021. Epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Infante, A.; Rodríguez, C.I. Cell and Cell-Free Therapies to Counteract Human Premature and Physiological Aging: MSCs Come to Light. J. Pers. Med. 2021, 11, 1043. https://doi.org/10.3390/jpm11101043
Infante A, Rodríguez CI. Cell and Cell-Free Therapies to Counteract Human Premature and Physiological Aging: MSCs Come to Light. Journal of Personalized Medicine. 2021; 11(10):1043. https://doi.org/10.3390/jpm11101043
Chicago/Turabian StyleInfante, Arantza, and Clara I. Rodríguez. 2021. "Cell and Cell-Free Therapies to Counteract Human Premature and Physiological Aging: MSCs Come to Light" Journal of Personalized Medicine 11, no. 10: 1043. https://doi.org/10.3390/jpm11101043