Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Stem Cell and Oxidative Stress-Inflammation Cycle

Author(s): Hatice Dogan Buzoglu, Ayse Burus, Yasemin Bayazıt and Michel Goldberg*

Volume 18, Issue 5, 2023

Published on: 16 November, 2022

Page: [641 - 652] Pages: 12

DOI: 10.2174/1574888X17666221012151425

Price: $65

conference banner
Abstract

Under a variety of physical and experimental settings, stem cells are able to self-renew and differentiate into specialized adult cells. MSCs (mesenchymal stromal/stem cells) are multipotent stem cells present in a wide range of fetal, embryonic, and adult tissues. They are the progenitors of a variety of specialized cells and are considered crucial tools in tissue engineering. MSCs, derived from various tissues, including cord blood, placenta, bone marrow, and dental tissues, have been extensively examined in tissue repair, immune modulation, etc. Increasing the vitality of MSCs and restoring cellular mechanisms are important factors in treatment success.

Oxidative stress harms cellular molecules such as DNA, proteins, and lipids due to the overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in cells and tissues or insufficiency of antioxidant systems that can inactivate them. Oxidative stress has a close link with inflammation as a pathophysiological process. ROS can mediate the expression of proinflammatory genes via intracellular signaling pathways and initiate the chronic inflammatory state. At the same time, inflammatory cells secrete a large number of reactive species that cause increased oxidative stress at sites of inflammation. In inflammatory diseases, the differentiation of stem cells and the regenerative and wound healing process can be affected differently by the increase of oxidative stress.

Recent studies have indicated that dental pulp stem cells (DPSCs), as a resource of adult stem cells, are an attractive option for cell therapy in diseases such as neurological diseases, diabetes, cardiological diseases, etc., as well as its treatment potential in pulp inflammation. The future of oxidative stressinflammation cycle and/or ageing therapies involves the selective elimination of senescent cells, also known as senolysis, which prevents various age-related diseases. Most pathologies are implicated on the effects of ageing without exerting undesirable side effects.

Graphical Abstract

[1]
Brignier AC, Gewirtz AM. Embryonic and adult stem cell therapy. J Allergy Clin Immunol 2010; 125(2): S336-44.
[http://dx.doi.org/10.1016/j.jaci.2009.09.032] [PMID: 20061008]
[2]
Baharvand H. Trends in stem cell biology and technology. New York, N.Y.: Humana Press 2009.
[http://dx.doi.org/10.1007/978-1-60327-905-5]
[3]
Gurusamy N, Alsayari A, Rajasingh S, Rajasingh J. Chapter one -adult stem cells for regenerative therapy. In: teplow DB, Ed. Progressin Molecular Biology and Translational Science 160. Massachusetts: Academic Press 2018; pp. 1-22.
[4]
Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal stem cells for regenerative medicine. Cells 2019; 8(8): 886.
[http://dx.doi.org/10.3390/cells8080886] [PMID: 31412678]
[5]
Parekkadan B, Milwid JM. Mesenchymal stem cells as therapeutics. Annu Rev Biomed Eng 2010; 12(1): 87-117.
[http://dx.doi.org/10.1146/annurev-bioeng-070909-105309] [PMID: 20415588]
[6]
Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968; 6(2): 230-47.
[http://dx.doi.org/10.1097/00007890-196803000-00009] [PMID: 5654088]
[7]
Li Q, Gao Z, Chen Y, Guan MX. The role of mitochondria in osteogenic, adipogenic and chondrogenic differentiation of mesenchymal stem cells. Protein Cell 2017; 8(6): 439-45.
[http://dx.doi.org/10.1007/s13238-017-0385-7] [PMID: 28271444]
[8]
Marquez CLA, Janowska WA, McGann LE, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology 2015; 71(2): 181-97.
[http://dx.doi.org/10.1016/j.cryobiol.2015.07.003] [PMID: 26186998]
[9]
Hocking DC. Therapeutic applications of extracellular matrix. Adv Wound Care 2015; 4(8): 441-3.
[http://dx.doi.org/10.1089/wound.2015.0652] [PMID: 26244100]
[10]
Mao X, Liu Y, Chen C, Shi S. Mesenchymal stem cells and their role in dental medicine. Dent Clin North Am 2017; 61(1): 161-72.
[http://dx.doi.org/10.1016/j.cden.2016.08.006] [PMID: 27912816]
[11]
Tsutsui T. Dental pulp stem cells: Advances to applications. Stem Cells Cloning 2020; 13: 33-42.
[http://dx.doi.org/10.2147/SCCAA.S166759] [PMID: 32104005]
[12]
Hofmann AD, Beyer M, Krause BU, Wobus M, Bornhäuser M, Rödel G. OXPHOS supercomplexes as a hallmark of the mitochondrial phenotype of adipogenic differentiated human MSCs. PLoS One 2012; 7(4): e35160.
[http://dx.doi.org/10.1371/journal.pone.0035160] [PMID: 22523573]
[13]
Hsu YC, Wu YT, Yu TH, Wei YH. Mitochondria in mesenchymal stem cell biology and cell therapy: From cellular differentiation to mitochondrial transfer. Semin Cell Dev Biol 2016; 52: 119-31.
[http://dx.doi.org/10.1016/j.semcdb.2016.02.011] [PMID: 26868759]
[14]
Tahara EB, Navarete FDT, Kowaltowski AJ. Tissue, substrate, and site-specific characteristics of mitochondrial reactive oxygen species generation. Free Radic Biol Med 2009; 46(9): 1283-97.
[http://dx.doi.org/10.1016/j.freeradbiomed.2009.02.008] [PMID: 19245829]
[15]
Pietilä M, Palomäki S, Lehtonen S, et al. Mitochondrial function and energy metabolism in umbilical cord blood and bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2012; 21(4): 575-88.
[http://dx.doi.org/10.1089/scd.2011.0023] [PMID: 21615273]
[16]
Varela RM, Embade N, Ariz U, Lu SC, Mato JM, Martínez CML. Non-alcoholic steatohepatitis and animal models: Understanding the human disease. Int J Biochem Cell Biol 2009; 41(5): 969-76.
[http://dx.doi.org/10.1016/j.biocel.2008.10.027] [PMID: 19027869]
[17]
Sart S, Song L, Li Y. Controlling redox status for stem cell survival, expansion, and differentiation. Oxid Med Cell Longev 2015; 2015: 105135.
[http://dx.doi.org/10.1155/2015/105135] [PMID: 26273419]
[18]
Wang W, Zhang Y, Lu W, Liu K. Mitochondrial reactive oxygen species regulate adipocyte differentiation of mesenchymal stem cells in hematopoietic stress induced by arabinosylcytosine. PLoS One 2015; 10(3): e0120629.
[http://dx.doi.org/10.1371/journal.pone.0120629] [PMID: 25768922]
[19]
Young IS, Woodside JV. Antioxidants in health and disease. J Clin Pathol 2001; 54(3): 176-86.
[http://dx.doi.org/10.1136/jcp.54.3.176] [PMID: 11253127]
[20]
Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ J 2012; 5(1): 9-19.
[http://dx.doi.org/10.1097/WOX.0b013e3182439613] [PMID: 23268465]
[21]
Kohlgrüber S, Upadhye A, Dyballa RN, McNamara CA, Altschmied J. Regulation of transcription factors by reactive oxygen species and nitric oxide in vascular physiology and pathology. Antioxid Redox Signal 2017; 26(13): 679-99.
[http://dx.doi.org/10.1089/ars.2016.6946] [PMID: 27841660]
[22]
Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. Clin Interv Aging 2018; 13: 757-72.
[http://dx.doi.org/10.2147/CIA.S158513] [PMID: 29731617]
[23]
Phaniendra A, Jestadi DB, Periyasamy L. Free radicals: properties, sources, targets, and their implication in various diseases. Indian J Clin Biochem 2015; 30(1): 11-26.
[http://dx.doi.org/10.1007/s12291-014-0446-0] [PMID: 25646037]
[24]
Lü JM, Lin PH, Yao Q, Chen C. Chemical and molecular mechanisms of antioxidants: Experimental approaches and model systems. J Cell Mol Med 2010; 14(4): 840-60.
[http://dx.doi.org/10.1111/j.1582-4934.2009.00897.x] [PMID: 19754673]
[25]
Pizzino G, Irrera N, Cucinotta M, et al. Oxidative stress: Harms and benefits for human health. Oxid Med Cell Longev 2017; 2017: 8416763.
[http://dx.doi.org/10.1155/2017/8416763] [PMID: 28819546]
[26]
Ping Z, Peng Y, Lang H, et al. Oxidative stress in radiation-induced cardiotoxicity. Oxid Med Cell Longev 2020; 2020: 3579143.
[http://dx.doi.org/10.1155/2020/3579143] [PMID: 32190171]
[27]
Ozcan M, Aydemir D, Bacanlı M, Anlar HG, Ulusu NN, Aksoy Y. Protective effects of antioxidant chlorophyllin in chemically induced breast cancer model in vivo. Biol Trace Elem Res 2021; 199(12): 4475-88.
[http://dx.doi.org/10.1007/s12011-021-02585-6] [PMID: 33624221]
[28]
Ozcan M, Esendagli G, Musdal Y, et al. Dual actions of the antioxidant chlorophyllin, a glutathione transferase P1-1 inhibitor, in tumorigenesis and tumor progression. J Cell Biochem 2018. Epub Ahead of Print
[PMID: 30484884]
[29]
Kim M, Kim C, Choi YS, Kim M, Park C, Suh Y. Age-related alterations in mesenchymal stem cells related to shift in differentiation from osteogenic to adipogenic potential: Implication to age-associated bone diseases and defects. Mech Ageing Dev 2012; 133(5): 215-25.
[http://dx.doi.org/10.1016/j.mad.2012.03.014] [PMID: 22738657]
[30]
Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: A review. Stem Cells Dev 2015; 24(10): 1150-63.
[http://dx.doi.org/10.1089/scd.2014.0484] [PMID: 25603196]
[31]
Rodrigues M, Turner O, Stolz D, Griffith LG, Wells A. Production of reactive oxygen species by multipotent stromal cells/mesenchymal stem cells upon exposure to fas ligand. Cell Transplant 2012; 21(10): 2171-87.
[http://dx.doi.org/10.3727/096368912X639035] [PMID: 22526333]
[32]
Lee DH, Lim BS, Lee YK, Yang HC. Effects of Hydrogen Peroxide (H2O2) on alkaline phosphatase activity and matrix mineralization of odontoblast and osteoblast cell lines. Cell Biol Toxicol 2006; 22(1): 39-46.
[http://dx.doi.org/10.1007/s10565-006-0018-z] [PMID: 16463018]
[33]
Marie PJ. Transcription factors controlling osteoblastogenesis. Arch Biochem Biophys 2008; 473(2): 98-105.
[http://dx.doi.org/10.1016/j.abb.2008.02.030] [PMID: 18331818]
[34]
Komori T. Signaling networks in RUNX2-dependent bone development. J Cell Biochem 2011; 112(3): 750-5.
[http://dx.doi.org/10.1002/jcb.22994] [PMID: 21328448]
[35]
Chen CT, Shih YRV, Kuo TK, Lee OK, Wei YH. Coordinated changes of mitochondrial biogenesis and antioxidant enzymes during osteogenic differentiation of human mesenchymal stem cells. Stem Cells 2008; 26(4): 960-8.
[http://dx.doi.org/10.1634/stemcells.2007-0509] [PMID: 18218821]
[36]
Tan J, Xu X, Tong Z, et al. Decreased osteogenesis of adult mesenchymal stem cells by reactive oxygen species under cyclic stretch: A possible mechanism of age related osteoporosis. Bone Res 2015; 3(1): 15003.
[http://dx.doi.org/10.1038/boneres.2015.3] [PMID: 26273536]
[37]
Yan H, Aziz E, Shillabeer G, et al. Nitric oxide promotes differentiation of rat white preadipocytes in culture. J Lipid Res 2002; 43(12): 2123-9.
[http://dx.doi.org/10.1194/jlr.M200305-JLR200] [PMID: 12454274]
[38]
Kanda Y, Hinata T, Kang SW, Watanabe Y. Reactive oxygen species mediate adipocyte differentiation in mesenchymal stem cells. Life Sci 2011; 89(7-8): 250-8.
[http://dx.doi.org/10.1016/j.lfs.2011.06.007] [PMID: 21722651]
[39]
Tormos KV, Anso E, Hamanaka RB, et al. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab 2011; 14(4): 537-44.
[http://dx.doi.org/10.1016/j.cmet.2011.08.007] [PMID: 21982713]
[40]
Higuchi M, Dusting GJ, Peshavariya H, et al. Differentiation of human adipose-derived stem cells into fat involves reactive oxygen species and Forkhead box O1 mediated upregulation of antioxidant enzymes. Stem Cells Dev 2013; 22(6): 878-88.
[http://dx.doi.org/10.1089/scd.2012.0306] [PMID: 23025577]
[41]
Nightingale H, Kemp K, Gray E, et al. Changes in expression of the antioxidant enzyme SOD3 occur upon differentiation of human bone marrow-derived mesenchymal stem cells in vitro. Stem Cells Dev 2012; 21(11): 2026-35.
[http://dx.doi.org/10.1089/scd.2011.0516] [PMID: 22132904]
[42]
Heywood HK, Lee DA. Bioenergetic reprogramming of articular chondrocytes by exposure to exogenous and endogenous reactive oxygen species and its role in the anabolic response to low oxygen. J Tissue Eng Regen Med 2017; 11(8): 2286-94.
[http://dx.doi.org/10.1002/term.2126] [PMID: 26799635]
[43]
Kim KS, Choi HW, Yoon HE, Kim IY. Reactive oxygen species generated by NADPH oxidase 2 and 4 are required for chondrogenic differentiation. J Biol Chem 2010; 285(51): 40294-302.
[http://dx.doi.org/10.1074/jbc.M110.126821] [PMID: 20952384]
[44]
Dayem AA, Choi HY, Kim JH, Cho SG. Role of oxidative stress in stem, cancer, and cancer stem cells. Cancers 2010; 2(2): 859-84.
[http://dx.doi.org/10.3390/cancers2020859] [PMID: 24281098]
[45]
Kim TH, Woo JS, Kim YK, Kim KH. Silibinin induces cell death through reactive oxygen species-dependent downregulation of notch-1/ERK/Akt signaling in human breast cancer cells. J Pharmacol Exp Ther 2014; 349(2): 268-78.
[http://dx.doi.org/10.1124/jpet.113.207563] [PMID: 24472723]
[46]
Shi Y, Hu Y, Lv C, Tu G. Effects of reactive oxygen species on differentiation of bone marrow mesenchymal stem cells. Ann Transplant 2016; 21: 695-700.
[http://dx.doi.org/10.12659/AOT.900463] [PMID: 27840405]
[47]
Yuan T, Yang T, Chen H, et al. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol 2019; 20: 247-60.
[http://dx.doi.org/10.1016/j.redox.2018.09.025] [PMID: 30384259]
[48]
Clark R, Kupper T. Old meets new: The interaction between innate and adaptive immunity. J Invest Dermatol 2005; 125(4): 629-37.
[http://dx.doi.org/10.1111/j.0022-202X.2005.23856.x] [PMID: 16185260]
[49]
Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010; 140(6): 805-20.
[http://dx.doi.org/10.1016/j.cell.2010.01.022] [PMID: 20303872]
[50]
Biswas SK. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxid Med Cell Longev 2016; 2016: 5698931.
[http://dx.doi.org/10.1155/2016/5698931] [PMID: 26881031]
[51]
Dröge W. Free radicals in the physiological control of cell function. Physiol Rev 2002; 82(1): 47-95.
[http://dx.doi.org/10.1152/physrev.00018.2001] [PMID: 11773609]
[52]
Popa WA, Mitran S, Sivanesan S, Chang E, Buga AM. ROS and brain diseases: The good, the bad, and the ugly. Oxid Med Cell Longev 2013; 2013: 963520.
[http://dx.doi.org/10.1155/2013/963520] [PMID: 24381719]
[53]
Salzano S, Checconi P, Hanschmann EM, et al. Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. Proc Natl Acad Sci USA 2014; 111(33): 12157-62.
[http://dx.doi.org/10.1073/pnas.1401712111] [PMID: 25097261]
[54]
Swirski FK, Nahrendorf M, Etzrodt M, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009; 325(5940): 612-6.
[http://dx.doi.org/10.1126/science.1175202] [PMID: 19644120]
[55]
Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol 2016; 142: 23-44.
[http://dx.doi.org/10.1016/j.pneurobio.2016.05.001] [PMID: 27166859]
[56]
Kim J, Hematti P. Mesenchymal stem cell–educated macrophages: A novel type of alternatively activated macrophages. Exp Hematol 2009; 37(12): 1445-53.
[http://dx.doi.org/10.1016/j.exphem.2009.09.004] [PMID: 19772890]
[57]
Villalta SA, Nguyen HX, Deng B, Gotoh T, Tidball JG. Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. Hum Mol Genet 2008; 18(3): 482-96.
[http://dx.doi.org/10.1093/hmg/ddn376] [PMID: 18996917]
[58]
Sehgal A, Donaldson DS, Pridans C, Sauter KA, Hume DA, Mabbott NA. The role of CSF1R-dependent macrophages in control of the intestinal stem-cell niche. Nat Commun 2018; 9(1): 1272.
[http://dx.doi.org/10.1038/s41467-018-03638-6] [PMID: 29593242]
[59]
Tidball JG. Inflammatory processes in muscle injury and repair. Am J Physiol Regul Integr Comp Physiol 2005; 288(2): R345-53.
[http://dx.doi.org/10.1152/ajpregu.00454.2004] [PMID: 15637171]
[60]
Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesenchymal stem cell-natural killer cell interactions: Evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 2006; 107(4): 1484-90.
[http://dx.doi.org/10.1182/blood-2005-07-2775] [PMID: 16239427]
[61]
Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005; 105(4): 1815-22.
[http://dx.doi.org/10.1182/blood-2004-04-1559] [PMID: 15494428]
[62]
Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: Implications in transplantation. Transplantation 2003; 75(3): 389-97.
[http://dx.doi.org/10.1097/01.TP.0000045055.63901.A9] [PMID: 12589164]
[63]
Wyles CC, Houdek MT, Behfar A, Sierra RJ. Mesenchymal stem cell therapy for osteoarthritis: Current perspectives. Stem Cells Cloning 2015; 8: 117-24.
[PMID: 26357483]
[64]
Denu RA, Hematti P. Effects of oxidative stress on mesenchymal stem cell biology. Oxid Med Cell Longev 2016; 2016: 2989076.
[http://dx.doi.org/10.1155/2016/2989076] [PMID: 27413419]
[65]
Herb M, Schramm M. Functions of ROS in macrophages and antimicrobial immunity. Antioxidants 2021; 10(2): 313.
[http://dx.doi.org/10.3390/antiox10020313] [PMID: 33669824]
[66]
Slauch JM. How does the oxidative burst of macrophages kill bacteria? Still an open question. Mol Microbiol 2011; 80(3): 580-3.
[http://dx.doi.org/10.1111/j.1365-2958.2011.07612.x] [PMID: 21375590]
[67]
Zimmerlin L, Park TS, Zambidis ET, Donnenberg VS, Donnenberg AD. Mesenchymal stem cell secretome and regenerative therapy after cancer. Biochimie 2013; 95(12): 2235-45.
[http://dx.doi.org/10.1016/j.biochi.2013.05.010] [PMID: 23747841]
[68]
Shi F, Guo LC, Zhu WD, et al. Human adipose tissue-derived MSCs improve psoriasis-like skin inflammation in mice by negatively regulating ROS. J Dermatolog Treat 2022; 33(4): 2129-36.
[http://dx.doi.org/10.1080/09546634.2021.1925622] [PMID: 34060412]
[69]
Rochette L, Mazini L, Malka G, Zeller M, Cottin Y, Vergely C. The crosstalk of Adipose-Derived Stem Cells (ADSC), oxidative stress, and inflammation in protective and adaptive responses. Int J Mol Sci 2020; 21(23): 9262.
[http://dx.doi.org/10.3390/ijms21239262] [PMID: 33291664]
[70]
Chen C, Tang Q, Zhang Y, et al. Metabolic reprogramming by HIF-1 activation enhances survivability of human adipose-derived stem cells in ischaemic microenvironments. Cell Prolif 2017; 50(5): e12363.
[http://dx.doi.org/10.1111/cpr.12363] [PMID: 28752896]
[71]
Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: Mechanisms and therapeutic potential. Trends Pharmacol Sci 2020; 41(9): 653-64.
[http://dx.doi.org/10.1016/j.tips.2020.06.009] [PMID: 32709406]
[72]
Philipp D, Suhr L, Wahlers T, Choi YH, Paunel GA. Preconditioning of bone marrow-derived mesenchymal stem cells highly strengthens their potential to promote IL-6-dependent M2b polarization. Stem Cell Res Ther 2018; 9(1): 286.
[http://dx.doi.org/10.1186/s13287-018-1039-2] [PMID: 30359316]
[73]
Zheng L, Teschler H, Guzman J, Hübner K, Striz I, Costabel U. Alveolar macrophage TNF-alpha release and BAL cell phenotypes in sarcoidosis. Am J Respir Crit Care Med 1995; 152(3): 1061-6.
[http://dx.doi.org/10.1164/ajrccm.152.3.7663784] [PMID: 7663784]
[74]
McClain CI, Hogden C, Nemeth K, et al. Bone marrow-derived Mesenchymal Stromal Cells (MSCs) modulate the inflammatory character of alveolar macrophages from sarcoidosis patients. J Clin Med 2020; 9(1): 278.
[http://dx.doi.org/10.3390/jcm9010278] [PMID: 31963936]
[75]
Mo IFY, Yip KHK, Chan WK, Law HKW, Lau YL, Chan GCF. Prolonged exposure to bacterial toxins downregulated expression of toll-like receptors in mesenchymal stromal cell-derived osteoprogenitors. BMC Cell Biol 2008; 9(1): 52.
[http://dx.doi.org/10.1186/1471-2121-9-52] [PMID: 18799018]
[76]
Chen GY, Shiah HC, Su HJ, et al. Baculovirus transduction of mesenchymal stem cells triggers the toll-like receptor 3 pathway. J Virol 2009; 83(20): 10548-56.
[http://dx.doi.org/10.1128/JVI.01250-09] [PMID: 19656899]
[77]
Lombardo E, DelaRosa O, Mancheño CP, Menta R, Ramírez C, Büscher D. Toll-like receptor-mediated signaling in human adipose-derived stem cells: Implications for immunogenicity and immunosuppressive potential. Tissue Eng Part A 2009; 15(7): 1579-89.
[http://dx.doi.org/10.1089/ten.tea.2008.0340] [PMID: 19061425]
[78]
Jeong SG, Cho GW. Endogenous ROS levels are increased in replicative senescence in human bone marrow mesenchymal stromal cells. Biochem Biophys Res Commun 2015; 460(4): 971-6.
[http://dx.doi.org/10.1016/j.bbrc.2015.03.136] [PMID: 25839657]
[79]
Zhang F, Peng W, Zhang J, et al. New strategy of bone marrow mesenchymal stem cells against oxidative stress injury via Nrf2 pathway: Oxidative stress preconditioning. J Cell Biochem 2019; 120(12): 19902-14.
[http://dx.doi.org/10.1002/jcb.29298] [PMID: 31347718]
[80]
Huang GTJ, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: Their biology and role in regenerative medicine. J Dent Res 2009; 88(9): 792-806.
[http://dx.doi.org/10.1177/0022034509340867] [PMID: 19767575]
[81]
Martens W, Bronckaers A, Politis C, Jacobs R, Lambrichts I. Dental stem cells and their promising role in neural regeneration: An update. Clin Oral Investig 2013; 17(9): 1969-83.
[http://dx.doi.org/10.1007/s00784-013-1030-3] [PMID: 23846214]
[82]
Feng J, Mantesso A, De Bari C, Nishiyama A, Sharpe PT. Dual origin of mesenchymal stem cells contributing to organ growth and repair. Proc Natl Acad Sci USA 2011; 108(16): 6503-8.
[http://dx.doi.org/10.1073/pnas.1015449108] [PMID: 21464310]
[83]
Kerkis I, Kerkis A, Dozortsev D, et al. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs 2006; 184(3-4): 105-16.
[http://dx.doi.org/10.1159/000099617] [PMID: 17409736]
[84]
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal human Dental Pulp Stem Cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci 2000; 97(25): 13625-30.
[http://dx.doi.org/10.1073/pnas.240309797] [PMID: 11087820]
[85]
Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res 2003; 18(4): 696-704.
[http://dx.doi.org/10.1359/jbmr.2003.18.4.696] [PMID: 12674330]
[86]
Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy 2006; 8(4): 315-7.
[http://dx.doi.org/10.1080/14653240600855905] [PMID: 16923606]
[87]
Karbanová J, Soukup T, Suchánek J, Pytlík R, Corbeil D, Mokrý J. Characterization of dental pulp stem cells from impacted third molars cultured in low serum-containing medium. Cells Tissues Organs 2011; 193(6): 344-65.
[http://dx.doi.org/10.1159/000321160] [PMID: 21071916]
[88]
Sakai K, Yamamoto A, Matsubara K, et al. Human dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat spinal cord by multiple neuro-regenerative mechanisms. J Clin Invest 2012; 122(1): 80-90.
[PMID: 22133879]
[89]
Shiba H, Fujita T, Doi N, et al. Differential effects of various growth factors and cytokines on the syntheses of DNA, type I collagen, laminin, fibronectin, osteonectin/Secreted Protein, Acidic And Rich In Cysteine (SPARC), and alkaline phosphatase by human pulp cells in culture. J Cell Physiol 1998; 174(2): 194-205.
[http://dx.doi.org/10.1002/(SICI)1097-4652(199802)174:2<194:AID-JCP7>3.0.CO;2-J] [PMID: 9428806]
[90]
Hwang HI, Lee TH, Jang YJ. Cell proliferation-inducing protein 52/mitofilin is a surface antigen on undifferentiated human dental pulp stem cells. Stem Cells Dev 2015; 24(11): 1309-19.
[http://dx.doi.org/10.1089/scd.2014.0387] [PMID: 25590652]
[91]
Wang L, Cheng L, Wang H, et al. Glycometabolic reprogramming associated with the initiation of human dental pulp stem cell differentiation. Cell Biol Int 2016; 40(3): 308-17.
[http://dx.doi.org/10.1002/cbin.10568] [PMID: 26634800]
[92]
Lee YH, Kang YM, Heo MJ, et al. The survival role of peroxisome proliferator-activated receptor gamma induces odontoblast differentiation against oxidative stress in human dental pulp cells. J Endod 2013; 39(2): 236-41.
[http://dx.doi.org/10.1016/j.joen.2012.11.006] [PMID: 23321237]
[93]
El Alami M, Viña AJ, Gambini J, et al. Activation of p38, p21, and NRF-2 mediates decreased proliferation of human dental pulp stem cells cultured under 21% O2. Stem Cell Reports 2014; 3(4): 566-73.
[http://dx.doi.org/10.1016/j.stemcr.2014.08.002] [PMID: 25358785]
[94]
Lv YJ, Yang Y, Sui BD, et al. Resveratrol counteracts bone loss via mitofilin-mediated osteogenic improvement of mesenchymal stem cells in senescence-accelerated mice. Theranostics 2018; 8(9): 2387-406.
[http://dx.doi.org/10.7150/thno.23620] [PMID: 29721087]
[95]
Sloan AJ, Waddington RJ. Dental pulp stem cells: What, where, how? Int J Paediatr Dent 2009; 19(1): 61-70.
[http://dx.doi.org/10.1111/j.1365-263X.2008.00964.x] [PMID: 19120509]
[96]
Alaidaroos NYA, Alraies A, Waddington RJ, Sloan AJ, Moseley R. Differential SOD2 and GSTZ1 profiles contribute to contrasting dental pulp stem cell susceptibilities to oxidative damage and premature senescence. Stem Cell Res Ther 2021; 12(1): 142.
[http://dx.doi.org/10.1186/s13287-021-02209-9] [PMID: 33596998]
[97]
Batouli S, Miura M, Brahim J, et al. Comparison of stem-cell-mediated osteogenesis and dentinogenesis. J Dent Res 2003; 82(12): 976-81.
[http://dx.doi.org/10.1177/154405910308201208] [PMID: 14630898]
[98]
Matsui M, Kobayashi T, Tsutsui TW. CD146 positive human dental pulp stem cells promote regeneration of dentin/pulp-like structures. Hum Cell 2018; 31(2): 127-38.
[http://dx.doi.org/10.1007/s13577-017-0198-2] [PMID: 29313241]
[99]
Prescott RS, Alsanea R, Fayad MI, et al. In vivo generation of dental pulp-like tissue by using dental pulp stem cells, a collagen scaffold, and dentin matrix protein 1 after subcutaneous transplantation in mice. J Endod 2008; 34(4): 421-6.
[http://dx.doi.org/10.1016/j.joen.2008.02.005] [PMID: 18358888]
[100]
Nakashima M, Akamine A. The application of tissue engineering to regeneration of pulp and dentin in endodontics. J Endod 2005; 31(10): 711-8.
[http://dx.doi.org/10.1097/01.don.0000164138.49923.e5] [PMID: 16186748]
[101]
Laird DJ, Von Andrian UH, Wagers AJ. Stem cell trafficking in tissue development, growth, and disease. Cell 2008; 132(4): 612-30.
[http://dx.doi.org/10.1016/j.cell.2008.01.041] [PMID: 18295579]
[102]
Galler KM, Widbiller M. Perspectives for cell-homing approaches to engineer dental pulp. J Endod 2017; 43(9): S40-5.
[http://dx.doi.org/10.1016/j.joen.2017.06.008] [PMID: 28778503]
[103]
Galvão I, Sugimoto MA, Vago JP, Machado MG, Sousa LP. Mediators of Inflammation. In: Riccardi C, Levi-Schaffer F, Tiligada E, Eds. Immunopharmacology and Inflammation. Cham: Springer International Publishing 2018; pp. 3-32.
[http://dx.doi.org/10.1007/978-3-319-77658-3_1]
[104]
Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008; 8(12): 958-69.
[http://dx.doi.org/10.1038/nri2448] [PMID: 19029990]
[105]
Cooper PR, Takahashi Y, Graham LW, Simon S, Imazato S, Smith AJ. Inflammation–regeneration interplay in the dentine–pulp complex. J Dent 2010; 38(9): 687-97.
[http://dx.doi.org/10.1016/j.jdent.2010.05.016] [PMID: 20580768]
[106]
Babb R, Chandrasekaran D, Carvalho MNV, Sharpe PT. Axin2-expressing cells differentiate into reparative odontoblasts via autocrine Wnt/β-catenin signaling in response to tooth damage. Sci Rep 2017; 7(1): 3102.
[http://dx.doi.org/10.1038/s41598-017-03145-6] [PMID: 28596530]
[107]
Neves VCM, Babb R, Chandrasekaran D, Sharpe PT. Promotion of natural tooth repair by small molecule GSK3 antagonists. Sci Rep 2017; 7(1): 39654.
[http://dx.doi.org/10.1038/srep39654] [PMID: 28067250]
[108]
Tsunawaki S, Sporn M, Ding A, Nathan C. Deactivation of macrophages by transforming growth factor-β. Nature 1988; 334(6179): 260-2.
[http://dx.doi.org/10.1038/334260a0] [PMID: 3041283]
[109]
Neves VCM, Yianni V, Sharpe PT. Macrophage modulation of dental pulp stem cell activity during tertiary dentinogenesis. Sci Rep 2020; 10(1): 20216.
[http://dx.doi.org/10.1038/s41598-020-77161-4] [PMID: 33214653]
[110]
Goldberg M, Farges J, Lacerdapinheiro S, et al. Inflammatory and immunological aspects of dental pulp repair. Pharmacol Res 2008; 58(2): 137-47.
[http://dx.doi.org/10.1016/j.phrs.2008.05.013] [PMID: 18602009]
[111]
Galler KM, Weber M, Korkmaz Y, Widbiller M, Feuerer M. Inflammatory response mechanisms of the dentine–pulp complex and the periapical tissues. Int J Mol Sci 2021; 22(3): 1480.
[http://dx.doi.org/10.3390/ijms22031480] [PMID: 33540711]
[112]
Zhang F, Lau SS, Monks TJ. The cytoprotective effect of N-acetyl-L-cysteine against ROS-induced cytotoxicity is independent of its ability to enhance glutathione synthesis. Toxicol Sci 2011; 120(1): 87-97.
[http://dx.doi.org/10.1093/toxsci/kfq364] [PMID: 21135414]
[113]
Jun SK, Yoon JY, Mahapatra C, et al. Ceria-incorporated MTA for accelerating odontoblastic differentiation via ROS downregulation. Dent Mater 2019; 35(9): 1291-9.
[http://dx.doi.org/10.1016/j.dental.2019.05.024] [PMID: 31255251]
[114]
Lee YH, Kim GE, Cho HJ, et al. Aging of in vitro pulp illustrates change of inflammation and dentinogenesis. J Endod 2013; 39(3): 340-5.
[http://dx.doi.org/10.1016/j.joen.2012.10.031] [PMID: 23402504]
[115]
Kirkland JL, Tchkonia T. Clinical strategies and animal models for developing senolytic agents. Exp Gerontol 2015; 68: 19-25.
[http://dx.doi.org/10.1016/j.exger.2014.10.012] [PMID: 25446976]
[116]
Baar MP, Brandt RMC, Putavet DA, et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 2017; 169(1): 132-147.e16.
[http://dx.doi.org/10.1016/j.cell.2017.02.031] [PMID: 28340339]
[117]
Van Deursen JM. Senolytic therapies for healthy longevity. Science 2019; 364(6441): 636-7.
[http://dx.doi.org/10.1126/science.aaw1299] [PMID: 31097655]
[118]
Kirkland JL, Tchkonia T. Senolytic drugs: From discovery to translation. J Intern Med 2020; 288(5): 518-36.
[http://dx.doi.org/10.1111/joim.13141] [PMID: 32686219]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy