1,726
Views
6
CrossRef citations to date
0
Altmetric
Research Article

Daphne mucronata enhances cell proliferation and protects human adipose stem cells against monosodium iodoacetate induced oxidative stress in vitro

ORCID Icon, , , &
Pages 495-508 | Received 15 Jun 2020, Accepted 15 Aug 2020, Published online: 31 Aug 2020

References

  • Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9:11–15.
  • Saparov A, Chen C-W, Beckman S, et al. The role of antioxidation and immunomodulation in postnatal multipotent stem cell-mediated cardiac repair. Int J Mol Sci. 2013;14:16258–16279.
  • Wang Y, Chen X, Cao W, et al. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol. 2014;15:1009.
  • Bunnell BA, Estes BT, Guilak F, et al. Differentiation of adipose stem cells. In: Adipose tissue protocols. Methods in Molecular Biology™. Humana Press; 2008. p. 155–171.
  • Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100:1249–1260.
  • Meliga E, Strem BM, Duckers H, et al. Adipose-derived cells. Cell Transplant. 2007;16:963–970.
  • Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–313.
  • Troyer DL, Weiss ML. Concise review: wharton’s jelly‐derived cells are a primitive stromal cell population. Stem Cells. 2008;26:591–599.
  • Weiss ML, Medicetty S, Bledsoe AR, et al. Human umbilical cord matrix stem cells: preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24:781–792.
  • Flynn A, Barry F, O’brien T. UC blood-derived mesenchymal stromal cells: an overview. Cytotherapy. 2007;9:717–726.
  • 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;8:315–317.
  • Chen Q, Shou P, Zheng C, et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 2016;23:1128.
  • Kokabu S, Lowery JW, Jimi E. Cell fate and differentiation of bone marrow mesenchymal stem cells. Stem Cells Int. 2016;2016:61–67.
  • Tao H, Han Z, Han ZC, et al. Proangiogenic features of mesenchymal stem cells and their therapeutic applications. Stem cells international. 2016. 2016.
  • Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell. 2015;17:11–22.
  • Gao F, Chiu S, Motan D, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7:e2062.
  • Amiri F, Jahanian-Najafabadi A, Roudkenar MH. In vitro augmentation of mesenchymal stem cells viability in stressful microenvironments. Cell Stress Chaperones. 2015;20:237–251.
  • Saparov A, Ogay V, Nurgozhin T, et al. Preconditioning of human mesenchymal stem cells to enhance their regulation of the immune response. In: Stem cells international. 2016. 2016.
  • Lee JH, Jung HK, Han YS, et al. Antioxidant effects of Cirsium setidens extract on oxidative stress in human mesenchymal stem cells. Mol Med Rep. 2016;14:3777–3784.
  • Sanap A, Chandravanshi B, Shah T, et al. Herbal pre-conditioning induces proliferation and delays senescence in Wharton’s Jelly mesenchymal stem cells. Biomed Pharmacother. 2017;93:772–778.
  • Di Giacomo C, Vanella L, Sorrenti V, et al. Effects of Tithonia diversifolia (Hemsl.) A. Gray extract on adipocyte differentiation of human mesenchymal stem cells. PloS One. 2015;10:e0122320.
  • Venkatesha SH, Berman BM, Moudgil KD. Herbal medicinal products target defined biochemical and molecular mediators of inflammatory autoimmune arthritis. Bioorg Med Chem. 2011;19:21–29.
  • Muluye RA, Bian Y, Alemu PN. Anti-inflammatory and antimicrobial effects of heat-clearing Chinese herbs: a current review. J Tradit Complement Med. 2014;4:93–98.
  • Ghosh S, May MJ, Kopp EB. NF-κB and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 1998;16:225–260.
  • Liu T, Zhang L, Joo D, et al. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.
  • Afzal MR, Haider HK, Idris NM, et al. Preconditioning promotes survival and angiomyogenic potential of mesenchymal stem cells in the infarcted heart via nf-κ b signaling. Antioxid Redox Signal. 2010;12:693–702.
  • Manu TM, Anand T, Pandareesh M, et al. Terminalia arjuna extract and arjunic acid mitigate cobalt chloride–induced hypoxia stress–mediated apoptosis in H9c2 cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 2019;392:1107–1119.
  • Kucukgul A, Isgor M, Duzguner V, et al. Antioxidant effects of oleuropein on hydrogen peroxide-induced neuronal stress-an in vitro study. Antiinflamm Antiallergy Agents Med Chem. 2020;19:74–84.
  • de Christo Scherer MM, Marques FM, Figueira MM, et al. Wound healing activity of terpinolene and α-phellandrene by attenuating inflammation and oxidative stress in vitro. J Tissue Viability. 2019;28:94–99.
  • Kim J-K, Park S-W, Kang J-W, et al. Effect of GCSB-5, a herbal formulation, on monosodium iodoacetate-induced osteoarthritis in rats. Evidence-Based Complementary and Alternative Medicine 2012; 2012.
  • Ra HJ, Oh MY, Kim HJ, et al. Effects of salmon DNA fraction in vitro and in a monosodium iodoacetate-induced osteoarthritis rat model. Korean J Physiol Pha. 2018;22:163–172.
  • Choi H-S, Im S, Park JW, et al. Protective effect of deer bone oil on cartilage destruction in rats with monosodium iodoacetate (MIA)-induced osteoarthritis. Biol Pharm Bull. 2016;39:2042–2051.
  • Lee YM, Son E, Kim S-H, et al. Effect of Alpinia oxyphylla extract in vitro and in a monosodium iodoacetate-induced osteoarthritis rat model. Phytomedicine. 2019;65:153095.
  • Jiang L, Li L, Geng C, et al. Monosodium iodoacetate induces apoptosis via the mitochondrial pathway involving ROS production and caspase activation in rat chondrocytes in vitro. J Orthop Res. 2013;31:364–369.
  • Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1005–L28.
  • Zaidi A, Bukhari SM, Khan FA, et al. Ethnobotanical, phytochemical and pharmacological aspects of Daphne mucronata (Thymeleaceae). Trop J Pharm Res. 2015;14:1517–1523.
  • Murad W, Ahmad A, Gilani SA, et al. Indigenous knowledge and folk use of medicinal plants by the tribal communities of Hazar Nao Forest, Malakand District, North Pakistan. J Med Plants Res. 2011;5:1072–1086.
  • Hamayun M. Traditional uses of some medicinal plants of Swat Valley. Indian J Tradition Knowl. 2007;6:636–641.
  • Khodadadian Z, Hassanpour-Ezatti M, Mousavi SZ, et al. Analgesic and anti-inflammatory potential of aerial parts of the Daphne mucronata Royle extract in mice: opioid-independent action. Asian Pac J Tropical Biomedicine. 2016;6:198–201.
  • Khan T, Abbasi BH, Khan MA, et al. Differential effects of thidiazuron on production of anticancer phenolic compounds in callus cultures of Fagonia indica. Appl Biochem Biotechnol. 2016;179:46–58.
  • Khan T, Abbasi BH, Zeb A, et al. Carbohydrate-induced biomass accumulation and elicitation of secondary metabolites in callus cultures of Fagonia indica. Ind Crops Prod. 2018;126:168–176.
  • Ozen T, Demirtas I, Aksit H. Determination of antioxidant activities of various extracts and essential oil compositions of Thymus praecox subsp. skorpilii var. skorpilii. Food Chem. 2011;124:58–64.
  • Carswell KA, Lee M-J, Fried SK Culture of isolated human adipocytes and isolated adipose tissue. In: Human cell culture protocols. Vol. 806. Humana Press; 2012. p. 203–214.
  • Naseer N, Bashir S, Latief N, et al. Human amniotic membrane as differentiating matrix for in vitro chondrogenesis. Regen Med. 2018;13:821–832.
  • Schachtele S, Clouser C, Aho J. Markers & methods to verify mesenchymal stem cell identity, potency, & quality. Mini R&D Syst. 2013; 10.
  • Rojewski MT, Weber BM, Schrezenmeier H. Phenotypic characterization of mesenchymal stem cells from various tissues. Transfusion Med Hemotherapy. 2008;35:168–184.
  • Zhao L, Hu C, Zhang P, et al. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med. 2019;23:720–730.
  • Li L, Chen X, Wang WE, et al. How to improve the survival of transplanted mesenchymal stem cell in ischemic heart? Stem Cells Int. 2016;2016:S139–S152.
  • Aravilli RK, Vikram SL, Kohila V. Phytochemicals as potential antidotes for targeting NF-κB in rheumatoid arthritis. 3 Biotech. 2017;7:253.
  • Pourreza N. Phenolic compounds as potential antioxidant. Jundishapur J Nat Pharm Prod. 2013;8:149.
  • Wang JM, Gu Y, Pan CJ, et al. Isolation, culture and identification of human adipose-derived stem cells. Exp Ther Med. 2017;13:1039–1043.
  • Dai K-R, Yan S-G, Yan W-Q, et al. Effects of naringin on the proliferation and osteogenic differentiation of human bone mesenchymal stem cell. Eur J Pharmacol. 2009;607:1–5.
  • Mahmoudi Z, Soleimani M, Saidi A, et al. Effect of Ferula gummosa ethanolic extract on osteogenesis in human mesenchymal stem cells. J Med Plant. 2013;2:50–59.
  • Li J, Zhou L, Tran HT, et al. Overexpression of laminin-8 in human dermal microvascular endothelial cells promotes angiogenesis-related functions. J Invest Dermatol. 2006;126:432–440.
  • Teplicki E, Ma Q, Castillo DE, et al. The effects of aloe vera on wound healing in cell proliferation, migration, and viability. Wounds. 2018;30:263–268.
  • Muniandy K, Gothai S, Tan WS, et al. In vitro wound healing potential of stem extract of alternanthera sessilis. Evidence-based complementary and alternative medicine. 2018. 2018.
  • Widowati W, Sardjono CT, Wijaya L, et al. Extract of Curcuma longa L. and (-)-Epigallo Catechin-3-Gallate enhanced proliferation of adipose tissue–derived mesenchymal stem cells (AD-MSCs) and differentiation of AD-MSCs into endothelial progenitor cells. J US China Med Sci. 2012;9:22–29.
  • Roughley PJ, Mort JS. The role of aggrecan in normal and osteoarthritic cartilage. J Exp Orthop. 2014;1:8.
  • Chen W, Cai S, Ren -Q-Q, et al. Recent advances in electrochemical sensing for hydrogen peroxide: a review. Analyst. 2012;137:49–58.
  • Sinha K, Das J, Pal PB, et al. Oxidative stress: the mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch Toxicol. 2013;87:1157–1180.
  • Weydert CJ, Cullen JJ. Measurement of superoxide dismutase, catalase and glutathione peroxidase in cultured cells and tissue. Nat Protoc. 2010;5:51.
  • Li W, Jiang B, Cao X, et al. Protective effect of lycopene on fluoride-induced ameloblasts apoptosis and dental fluorosis through oxidative stress-mediated Caspase pathways. Chem Biol Interact. 2017;261:27–34.
  • Wu L, Huang X, Li L, et al. Insights on biology and pathology of HIF-1α/-2α, TGFα/BMP, Wnt/β-catenin, and NF-κB pathways in osteoarthritis. Curr Pharm Des. 2012;18:3293–3312.
  • Lawrence T. The nuclear factor NF-κB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1:a001651.
  • Li Q, Verma IM. NF-κB regulation in the immune system. Nat Rev Immunol. 2002;2:725–734.
  • Udalova IA, Mott R, Field D, et al. Quantitative prediction of NF-κB DNA–protein interactions. PNAS 2002; 99:8167–8172.
  • Xu W, Huang M, Zhang Y, et al. Extracts of Bauhinia championii (Benth.) Benth. inhibit NF-< kappa> B-signaling in a rat model of collagen-induced arthritis and primary synovial cells. J Ethnopharmacol. 2016;185:140–146.
  • Oliviero M, Romilde I, Beatrice MM, et al. Evaluations of thyme extract effects in human normal bronchial and tracheal epithelial cell lines and in human lung cancer cell line. Chem Biol Interact. 2016;256:125–133.
  • Israël A. The IKK complex, a central regulator of NF-κB activation. Cold Spring Harb Perspect Biol. 2010;2:a000158.
  • Paul A, Edwards J, Pepper C, et al. Inhibitory-Κb kinase (IKK) α and nuclear factor-κB (NFκB)-inducing kinase (NIK) as anti-cancer drug targets. Cells. 2018;7:176.
  • Liu F, Xia Y, Parker AS, et al. IKK biology. Immunol Rev. 2012;246:239–253.
  • Jeong YE, Lee M-Y. Anti-inflammatory activity of Populus deltoides leaf extract via modulating NF-κB and p38/JNK pathways. Int J Mol Sci. 2018;19:3746.
  • Yamamoto Y, Gaynor RB. IκB kinases: key regulators of the NF-κB pathway. Trends Biochem Sci. 2004;29:72–79.
  • Ammon H. Modulation of the immune system by Boswellia serrata extracts and boswellic acids. Phytomedicine. 2010;17:862–867.
  • Luetragoon T, Pankla Sranujit R, Noysang C, et al. Bioactive Compounds in Moringa oleifera Lam. Leaves Inhibit the Pro-Inflammatory Mediators in Lipopolysaccharide-Induced Human Monocyte-Derived Macrophages. Molecules. 2020;25:191.
  • Inada M, Wang Y, Byrne MH, et al. Critical roles for collagenase-3 (Mmp13) in development of growth plate cartilage and in endochondral ossification. PNAS 2004; 101:17192–17197.
  • Kudo Y, Iizuka S, Yoshida M, et al. Matrix metalloproteinase-13 (MMP-13) directly and indirectly promotes tumor angiogenesis. J Biol Chem. 2012;287:38716–38728.
  • Manicone AM, McGuire JK Matrix metalloproteinases as modulators of inflammation. Semin Cell Dev Biol. Elsevier. 2008;19:34–41.
  • Vandenbroucke RE, Dejonckheere E, Van Hauwermeiren F, et al. Matrix metalloproteinase 13 modulates intestinal epithelial barrier integrity in inflammatory diseases by activating TNF. EMBO Mol Med. 2013;5:1000–1016.
  • Schönbeck U, Mach F, Libby P. Generation of biologically active IL-1β by matrix metalloproteinases: a novel caspase-1-independent pathway of IL-1β processing. J Immunol. 1998;161:3340–3346.
  • Phienwej H, Swasdichira I-S, Amnuoypol S, et al. Tinospora crispa extract inhibits MMP-13 and migration of head and neck squamous cell carcinoma cell lines. Asian Pac J Tropical Biomedicine. 2015;5:738–743.
  • Wongwat T, Srihaphon K, Pitaksutheepong C, et al. Suppression of inflammatory mediators and matrix metalloproteinase (MMP)-13 by Morus alba stem extract and oxyresveratrol in RAW 264.7 cells and C28/I2 human chondrocytes. J Tradit Complement Med. 2020;10:132–140.