Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

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

Review Article

Mesenchymal Stem Cells and their Exosomes: Promising Therapeutics for Chronic Pain

Author(s): Jinxuan Ren, Na Liu, Na Sun, Kehan Zhang and Lina Yu*

Volume 14, Issue 8, 2019

Page: [644 - 653] Pages: 10

DOI: 10.2174/1574888X14666190912162504

Price: $65

Abstract

Chronic pain is a common condition that seriously affects the quality of human life with variable etiology and complicated symptoms; people who suffer from chronic pain may experience anxiety, depression, insomnia, and other harmful emotions. Currently, chronic pain treatments are nonsteroidal anti-inflammatory drugs and opioids; these drugs are demonstrated to be insufficient and cause severe side effects. Therefore, research into new therapeutic strategies for chronic pain is a top priority. In recent years, stem cell transplantation has been demonstrated to be a potent alternative for the treatment of chronic pain. Mesenchymal stem cells (MSCs), a type of pluripotent stem cell, exhibit multi-directional differentiation, promotion of stem cell implantation, and immune regulation; they have also been shown to exert analgesic effects in several chronic pain models. Exosomes produced by MSCs have been demonstrated to relieve painful symptoms with fewer side effects. In this review, we summarize the therapeutic use of MSCs in various chronic pain studies. We also discuss ways to enhance the treatment effect of MSCs. We predict in the future, cell-free therapies for chronic pain will develop from exosomes secreted by MSCs.

Keywords: Chronic pain, mesenchymal stem cells, exosomes, analgesia, cytokines, cell-free.

[1]
Uttam S, Wong C, Amorim IS, et al. Translational profiling of dorsal root ganglia and spinal cord in a mouse model of neuropathic pain. Neurobiol Pain 2018; 4: 35-44.
[http://dx.doi.org/10.1016/j.ynpai.2018.04.001] [PMID: 30906902]
[2]
Davis DA, Ghantous ME, Farmer MA, Baria AT, Apkarian AV. Identifying brain nociceptive information transmission in patients with chronic somatic pain. Pain Rep 2016; 1(4)e575
[http://dx.doi.org/10.1097/PR9.0000000000000575] [PMID: 28503674]
[3]
Zhou XL, Zhang CJ, Peng YN, Wang Y, Xu HJ, Liu CM. The receptor tyrosine kinase-like orphan receptor ROR2 modulates neuropathic pain via phosphorylation of N-methyl-D-aspartate receptor subunit GluN2B. Br J Anaesth 2018.
[4]
Gerdle B, Åkerblom S, Brodda Jansen G, et al. Who benefits from multimodal rehabilitation - an exploration of pain, psychological distress, and life impacts in over 35,000 chronic pain patients identified in the swedish quality registry for pain rehabilitation. J Pain Res 2019; 12: 891-908.
[http://dx.doi.org/10.2147/JPR.S190003] [PMID: 30881099]
[5]
Huh Y, Ji RR, Chen G. Neuroinflammation, Bone Marrow Stem Cells, and Chronic Pain. Front Immunol 2017; 8: 1014.
[http://dx.doi.org/10.3389/fimmu.2017.01014] [PMID: 28871264]
[6]
Tsang A, Von Korff M, Lee S, et al. Common chronic pain conditions in developed and developing countries: Gender and age differences and comorbidity with depression-anxiety disorders. J Pain 2008; 9(10): 883-91.
[http://dx.doi.org/10.1016/j.jpain.2008.05.005] [PMID: 18602869]
[7]
Mayer S, Spickschen J, Stein KV, Crevenna R, Dorner TE, Simon J. The societal costs of chronic pain and its determinants: The case of Austria. PLoS One 2019; 14(3)e0213889
[http://dx.doi.org/10.1371/journal.pone.0213889] [PMID: 30893370]
[8]
Gaskin DJ, Richard P. The economic costs of pain in the United States. J Pain 2012; 13(8): 715-24.
[http://dx.doi.org/10.1016/j.jpain.2012.03.009] [PMID: 22607834]
[9]
King S, Chambers CT, Huguet A, et al. The epidemiology of chronic pain in children and adolescents revisited: A systematic review. Pain 2011; 152(12): 2729-38.
[http://dx.doi.org/10.1016/j.pain.2011.07.016] [PMID: 22078064]
[10]
Ji RR, Xu ZZ, Gao YJ. Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discov 2014; 13(7): 533-48.
[http://dx.doi.org/10.1038/nrd4334] [PMID: 24948120]
[11]
Hosseini M, Yousefifard M, Aziznejad H, Nasirinezhad F. The effect of bone marrow-derived mesenchymal stem cell transplantation on allodynia and hyperalgesia in neuropathic animals: A systematic review with meta-analysis. Biol Blood Marrow Transplant 2015; 21(9): 1537-44.
[http://dx.doi.org/10.1016/j.bbmt.2015.05.008] [PMID: 25985918]
[12]
Liu L, Hua Z, Shen J, et al. Comparative efficacy of multiple variables of mesenchymal stem cell transplantation for the treatment of neuropathic pain in rats. Mil Med 2017; 182(S1): 175-84.
[http://dx.doi.org/10.7205/MILMED-D-16-00096] [PMID: 28291470]
[13]
Guo W, Wang H, Zou S, et al. Bone marrow stromal cells produce long-term pain relief in rat models of persistent pain. Stem Cells 2011; 29(8): 1294-303.
[http://dx.doi.org/10.1002/stem.667] [PMID: 21630378]
[14]
Yousefifard M, Nasirinezhad F, Shardi Manaheji H, Janzadeh A, Hosseini M, Keshavarz M. Human bone marrow-derived and umbilical cord-derived mesenchymal stem cells for alleviating neuropathic pain in a spinal cord injury model. Stem Cell Res Ther 2016; 7: 36.
[http://dx.doi.org/10.1186/s13287-016-0295-2] [PMID: 26957122]
[15]
Brini AT, Amodeo G, Ferreira LM, et al. Therapeutic effect of human adipose-derived stem cells and their secretome in experimental diabetic pain. Sci Rep 2017; 7(1): 9904.
[http://dx.doi.org/10.1038/s41598-017-09487-5] [PMID: 28851944]
[16]
Hurley ET, Yasui Y, Gianakos AL, et al. Limited evidence for adipose-derived stem cell therapy on the treatment of osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2018; 26(11): 3499-507.
[http://dx.doi.org/10.1007/s00167-018-4955-x] [PMID: 29713784]
[17]
Heidari B, Shirazi A, Akhondi MM, et al. Comparison of proliferative and multilineage differentiation potential of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tissue. Avicenna J Med Biotechnol 2013; 5(2): 104-17.
[PMID: 23799179]
[18]
Tomasello L, Mauceri R, Coppola A, et al. Mesenchymal stem cells derived from inflamed dental pulpal and gingival tissue: a potential application for bone formation. Stem Cell Res Ther 2017; 8(1): 179.
[http://dx.doi.org/10.1186/s13287-017-0633-z] [PMID: 28764802]
[19]
Xie J, Liu B, Chen J, et al. Umbilical cord-derived mesenchymal stem cells alleviated inflammation and inhibited apoptosis in interstitial cystitis via AKT/mTOR signaling pathway. Biochem Biophys Res Commun 2018; 495(1): 546-52.
[http://dx.doi.org/10.1016/j.bbrc.2017.11.072] [PMID: 29137981]
[20]
Gullo F, De Bari C. Prospective purification of a subpopulation of human synovial mesenchymal stem cells with enhanced chondro-osteogenic potency. Rheumatology (Oxford) 2013; 52(10): 1758-68.
[http://dx.doi.org/10.1093/rheumatology/ket205] [PMID: 23804221]
[21]
Zhang Y, Zhang W, Wang H, Yang B. miR-21 contributes to human amniotic membrane-derived mesenchymal stem cell growth and human amniotic membrane-derived mesenchymal stem cell-induced immunoregulation. Genet Test Mol Biomarkers 2018; 22(12): 665-73.
[http://dx.doi.org/10.1089/gtmb.2018.0116] [PMID: 30481073]
[22]
Seo BM, Miura M, Gronthos S, et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004; 364(9429): 149-55.
[http://dx.doi.org/10.1016/S0140-6736(04)16627-0] [PMID: 15246727]
[23]
Fei X, Jiang S, Zhang S, et al. Isolation, culture, and identification of amniotic fluid-derived mesenchymal stem cells. Cell Biochem Biophys 2013; 67(2): 689-94.
[http://dx.doi.org/10.1007/s12013-013-9558-z] [PMID: 23508888]
[24]
Meier RP, Müller YD, Morel P, Gonelle-Gispert C, Bühler LH. Transplantation of mesenchymal stem cells for the treatment of liver diseases, is there enough evidence? Stem Cell Res (Amst) 2013; 11(3): 1348-64.
[http://dx.doi.org/10.1016/j.scr.2013.08.011] [PMID: 24090934]
[25]
Shi Y, Wang Y, Li Q, et al. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat Rev Nephrol 2018; 14(8): 493-507.
[http://dx.doi.org/10.1038/s41581-018-0023-5] [PMID: 29895977]
[26]
Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell 2011; 9(1): 11-5.
[http://dx.doi.org/10.1016/j.stem.2011.06.008] [PMID: 21726829]
[27]
Chen G, Park CK, Xie RG, Ji RR. Intrathecal bone marrow stromal cells inhibit neuropathic pain via TGF-β secretion. J Clin Invest 2015; 125(8): 3226-40.
[http://dx.doi.org/10.1172/JCI80883] [PMID: 26168219]
[28]
Iwasa T, Afroz S, Inoue M, et al. IL-10 and CXCL2 in trigeminal ganglia in neuropathic pain. Neurosci Lett 2019; 703: 132-8.
[http://dx.doi.org/10.1016/j.neulet.2019.03.031] [PMID: 30904573]
[29]
Fischer G, Wang F, Xiang H, Bai X, Yu H, Hogan QH. Inhibition of neuropathic hyperalgesia by intrathecal bone marrow stromal cells is associated with alteration of multiple soluble factors in cerebrospinal fluid. Exp Brain Res 2017; 235(9): 2627-38.
[http://dx.doi.org/10.1007/s00221-017-5000-x] [PMID: 28573310]
[30]
Galindo S, Herreras JM, López-Paniagua M, et al. Therapeutic effect of human adipose tissue-derived mesenchymal stem cells in experimental corneal failure due to limbal stem cell niche damage. Stem Cells 2017; 35(10): 2160-74.
[http://dx.doi.org/10.1002/stem.2672] [PMID: 28758321]
[31]
Shiue SJ, Rau RH, Shiue HS, et al. Mesenchymal stem cell exosomes as a cell-free therapy for nerve injury-induced pain in rats. Pain 2019; 160(1): 210-23.
[http://dx.doi.org/10.1097/j.pain.0000000000001395] [PMID: 30188455]
[32]
Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci 2014; 15(3): 4142-57.
[http://dx.doi.org/10.3390/ijms15034142] [PMID: 24608926]
[33]
Toh WS, Lai RC, Zhang B, Lim SK. MSC exosome works through a protein-based mechanism of action. Biochem Soc Trans 2018; 46(4): 843-53.
[http://dx.doi.org/10.1042/BST20180079] [PMID: 29986939]
[34]
Yang Y, Ye Y, Su X, He J, Bai W, He X. MSCs-Derived Exosomes and Neuroinflammation, Neurogenesis and Therapy of Traumatic Brain Injury. Front Cell Neurosci 2017; 11: 55.
[http://dx.doi.org/10.3389/fncel.2017.00055] [PMID: 28293177]
[35]
Knopp KL, Simmons RMA, Guo W, et al. Modulation of TARP γ8-Containing AMPA Receptors as a Novel Therapeutic Approach for Chronic Pain. J Pharmacol Exp Ther 2019; 369(3): 345-63.
[http://dx.doi.org/10.1124/jpet.118.250126] [PMID: 30910921]
[36]
Marin-Bañasco C, Benabdellah K, Melero-Jerez C, et al. Gene therapy with mesenchymal stem cells expressing IFN-ß ameliorates neuroinflammation in experimental models of multiple sclerosis. Br J Pharmacol 2017; 174(3): 238-53.
[http://dx.doi.org/10.1111/bph.13674] [PMID: 27882538]
[37]
Mathot F, Shin AY, Van Wijnen AJ. Targeted stimulation of MSCs in peripheral nerve repair. Gene 2019; 710: 17-23.
[http://dx.doi.org/10.1016/j.gene.2019.02.078] [PMID: 30849542]
[38]
Saldaña L, Bensiamar F, Vallés G, Mancebo FJ, García-Rey E, Vilaboa N. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther 2019; 10(1): 58.
[http://dx.doi.org/10.1186/s13287-019-1156-6] [PMID: 30760316]
[39]
Putra A, Ridwan FB, Putridewi AI, et al. The Role of TNF-α induced MSCs on Suppressive Inflammation by Increasing TGF-β and IL-10. Open Access Maced J Med Sci 2018; 6(10): 1779-83.
[http://dx.doi.org/10.3889/oamjms.2018.404] [PMID: 30455748]
[40]
Rosado IR, Carvalho PH, Alves EG, et al. Immunomodulatory and neuroprotective effect of cryopreserved allogeneic mesenchymal stem cells on spinal cord injury in rats. Genet Mol Res 2017; 16(1)
[http://dx.doi.org/10.4238/gmr16019555] [PMID: 28340277]
[41]
Ichiseki T, Shimazaki M, Ueda Y, et al. Intraarticularly-injected mesenchymal stem cells stimulate anti-inflammatory molecules and inhibit pain related protein and chondrolytic enzymes in a monoiodoacetate-induced rat arthritis model. Int J Mol Sci 2018; 19(1)E203
[http://dx.doi.org/10.3390/ijms19010203] [PMID: 29315262]
[42]
Gupta PK, Chullikana A, Rengasamy M, 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(1): 301.
[http://dx.doi.org/10.1186/s13075-016-1195-7] [PMID: 27993154]
[43]
Lee SY, Lee SH, Na HS, et al. The therapeutic effect of STAT3 signaling-suppressed MSC on pain and articular cartilage damage in a rat model of monosodium iodoacetate-induced osteoarthritis. Front Immunol 2018; 9: 2881.
[http://dx.doi.org/10.3389/fimmu.2018.02881] [PMID: 30619261]
[44]
Gold MS, Gebhart GF. Nociceptor sensitization in pain pathogenesis. Nat Med 2010; 16(11): 1248-57.
[http://dx.doi.org/10.1038/nm.2235] [PMID: 20948530]
[45]
Batista CM, Mariano ED, Onuchic F, et al. Characterization of traumatic spinal cord injury model in relation to neuropathic pain in the rat. Somatosens Mot Res 2019; 36(1): 14-23.
[http://dx.doi.org/10.1080/08990220.2018.1563537] [PMID: 30870070]
[46]
Haanpää M, Attal N, Backonja M, et al. NeuPSIG guidelines on neuropathic pain assessment. Pain 2011; 152(1): 14-27.
[http://dx.doi.org/10.1016/j.pain.2010.07.031] [PMID: 20851519]
[47]
van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: A systematic review of epidemiological studies. Pain 2014; 155(4): 654-62.
[http://dx.doi.org/10.1016/j.pain.2013.11.013] [PMID: 24291734]
[48]
Lindsay SL, Toft A, Griffin JMM, Emraja A, Barnett SC, Riddell JS. Human olfactory mesenchymal stromal cell transplants promote remyelination and earlier improvement in gait co-ordination after spinal cord injury. Glia 2017; 65(4): 639-56.
[http://dx.doi.org/10.1002/glia.23117] [PMID: 28144983]
[49]
Qu J, Zhang H. Roles of Mesenchymal Stem Cells in Spinal Cord Injury. Stem Cells Int 2017.20175251313
[http://dx.doi.org/10.1155/2017/5251313] [PMID: 28630630]
[50]
Han D, Wu C, Xiong Q, Zhou L, Tian Y. Anti-inflammatory mechanism of bone marrow mesenchymal stem cell transplantation in rat model of spinal cord injury. Cell Biochem Biophys 2015; 71(3): 1341-7.
[http://dx.doi.org/10.1007/s12013-014-0354-1] [PMID: 25388837]
[51]
Roh DH, Seo MS, Choi HS, et al. Transplantation of human umbilical cord blood or amniotic epithelial stem cells alleviates mechanical allodynia after spinal cord injury in rats. Cell Transplant 2013; 22(9): 1577-90.
[http://dx.doi.org/10.3727/096368912X659907] [PMID: 23294734]
[52]
Shin DA, Pennant WA, Yoon DH, Ha Y, Kim KN. Co-transplantation of bone marrow-derived mesenchymal stem cells and nanospheres containing FGF-2 improve cell survival and neurological function in the injured rat spinal cord. Acta Neurochir (Wien) 2014; 156(2): 297-303.
[http://dx.doi.org/10.1007/s00701-013-1963-y] [PMID: 24352373]
[53]
Cizkova D, Novotna I, Slovinska L, et al. Repetitive intrathecal catheter delivery of bone marrow mesenchymal stromal cells improves functional recovery in a rat model of contusive spinal cord injury. J Neurotrauma 2011; 28(9): 1951-61.
[http://dx.doi.org/10.1089/neu.2010.1413] [PMID: 20822464]
[54]
Osaka M, Honmou O, Murakami T, et al. Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res 2010; 1343: 226-35.
[http://dx.doi.org/10.1016/j.brainres.2010.05.011] [PMID: 20470759]
[55]
Dos Santos Ramalho B, Marques Pestana F, Andrade Prins C, et al. Effects of different doses of mesenchymal stem cells on functional recovery after compressive spinal-cord injury in mice. Neuroscience 2019; 400: 17-32.
[http://dx.doi.org/10.1016/j.neuroscience.2018.12.005] [PMID: 30553796]
[56]
Yang C, Wang G, Ma F, et al. Repeated injections of human umbilical cord blood-derived mesenchymal stem cells significantly promotes functional recovery in rabbits with spinal cord injury of two noncontinuous segments. Stem Cell Res Ther 2018; 9(1): 136.
[http://dx.doi.org/10.1186/s13287-018-0879-0] [PMID: 29751769]
[57]
Lee MJ, Yoon TG, Kang M, Kim HJ, Kang KS. Effect of subcutaneous treatment with human umbilical cord blood-derived multipotent stem cells on peripheral neuropathic pain in rats. Korean J Physiol Pharmacol 2017; 21(2): 153-60.
[http://dx.doi.org/10.4196/kjpp.2017.21.2.153] [PMID: 28280408]
[58]
Mao F, Xu WR, Qian H, et al. Immunosuppressive effects of mesenchymal stem cells in collagen-induced mouse arthritis. Inflamm Res 2010; 59(3): 219-25.
[http://dx.doi.org/10.1007/s00011-009-0090-y] [PMID: 19763787]
[59]
Schäfer S, Berger JV, Deumens R, Goursaud S, Hanisch UK, Hermans E. Influence of intrathecal delivery of bone marrow-derived mesenchymal stem cells on spinal inflammation and pain hypersensitivity in a rat model of peripheral nerve injury. J Neuroinflammation 2014; 11: 157.
[http://dx.doi.org/10.1186/s12974-014-0157-8] [PMID: 25212534]
[60]
Evangelista AF, Vannier-Santos MA, de Assis Silva GS, et al. Bone marrow-derived mesenchymal stem/stromal cells reverse the sensorial diabetic neuropathy via modulation of spinal neuroinflammatory cascades. J Neuroinflammation 2018; 15(1): 189.
[http://dx.doi.org/10.1186/s12974-018-1224-3] [PMID: 29933760]
[61]
Sun Y, Tian Y, Li H, Zhang D, Sun Q. Antinociceptive effect of intrathecal injection of genetically engineered human bone marrow stem cells expressing the human proenkephalin gene in a rat model of bone cancer pain. Pain Res Manag 2017.20177346103
[http://dx.doi.org/10.1155/2017/7346103] [PMID: 28286408]
[62]
Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: An update. Cell Transplant 2016; 25(5): 829-48.
[http://dx.doi.org/10.3727/096368915X689622] [PMID: 26423725]
[63]
Xu P, Yang X. The Efficacy and Safety of Mesenchymal Stem Cell Transplantation for Spinal Cord Injury Patients: A Meta-Analysis and Systematic Review. Cell Transplant 2019; 28(1): 36-46.
[http://dx.doi.org/10.1177/0963689718808471] [PMID: 30362373]
[64]
Vaquero J, Zurita M, Rico MA, et al. Neurological Cell Therapy Group. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy 2017; 19(3): 349-59.
[http://dx.doi.org/10.1016/j.jcyt.2016.12.002] [PMID: 28089079]
[65]
Vaquero J, Zurita M, Rico MA, et al. Neurological Cell Therapy Group from Puerta de Hierro-Majadahonda Hospital. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: Safety and efficacy of the 100/3 guideline. Cytotherapy 2018; 20(6): 806-19.
[http://dx.doi.org/10.1016/j.jcyt.2018.03.032] [PMID: 29853256]
[66]
Vaquero J, Zurita M, Rico MA, et al. Intrathecal administration of autologous bone marrow stromal cells improves neuropathic pain in patients with spinal cord injury. Neurosci Lett 2018; 670: 14-8.
[http://dx.doi.org/10.1016/j.neulet.2018.01.035] [PMID: 29366770]
[67]
Liu J, Han D, Wang Z, et al. Clinical analysis of the treatment of spinal cord injury with umbilical cord mesenchymal stem cells. Cytotherapy 2013; 15(2): 185-91.
[http://dx.doi.org/10.1016/j.jcyt.2012.09.005] [PMID: 23321330]
[68]
Oh SK, Choi KH, Yoo JY, Kim DY, Kim SJ, Jeon SR. A Phase III clinical trial showing limited efficacy of autologous mesenchymal stem cell therapy for spinal cord injury. Neurosurgery 2016; 78(3): 436-47.
[http://dx.doi.org/10.1227/NEU.0000000000001056] [PMID: 26891377]
[69]
Zhao Y, Tang F, Xiao Z, et al. Clinical study of neuro regen scaffold combined with human mesenchymal stem cells for the repair of chronic complete spinal cord injury. Cell Transplant 2017; 26(5): 891-900.
[http://dx.doi.org/10.3727/096368917X695038] [PMID: 28185615]
[70]
Orozco L, Munar A, Soler R, et al. Treatment of knee osteoarthritis with autologous mesenchymal stem cells: a pilot study. Transplantation 2013; 95(12): 1535-41.
[http://dx.doi.org/10.1097/TP.0b013e318291a2da] [PMID: 23680930]
[71]
Chahal J, Gómez-Aristizábal A, Shestopaloff K, et al. Bone marrow mesenchymal stromal cells in patients with osteoarthritis results in overall improvement in pain and symptoms and reduces synovial inflammation. Stem Cells Transl Med 2019.
[http://dx.doi.org/10.1002/sctm.18-0183] [PMID: 30964245]
[72]
Lee WS, Kim HJ, Kim KI, Kim GB, Jin W. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: A Phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med 2019; 8(6): 504-11.
[http://dx.doi.org/10.1002/sctm.18-0122] [PMID: 30835956]
[73]
Matas J, Orrego M, Amenabar D, 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(3): 215-24.
[http://dx.doi.org/10.1002/sctm.18-0053] [PMID: 30592390]
[74]
Elabd C, Centeno CJ, Schultz JR, Lutz G, Ichim T, Silva FJ. Intra-discal injection of autologous, hypoxic cultured bone marrow-derived mesenchymal stem cells in five patients with chronic lower back pain: a long-term safety and feasibility study. J Transl Med 2016; 14: 253.
[http://dx.doi.org/10.1186/s12967-016-1015-5] [PMID: 27585696]
[75]
Centeno C, Markle J, Dodson E, et al. Treatment of lumbar degenerative disc disease-associated radicular pain with culture-expanded autologous mesenchymal stem cells: a pilot study on safety and efficacy. J Transl Med 2017; 15(1): 197.
[http://dx.doi.org/10.1186/s12967-017-1300-y] [PMID: 28938891]
[76]
Pang X, Yang H, Peng B. Human umbilical cord mesenchymal stem cell transplantation for the treatment of chronic discogenic low back pain. Pain Physician 2014; 17(4): E525-30.
[PMID: 25054402]
[77]
Kumar H, Ha DH, Lee EJ, et al. Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study. Stem Cell Res Ther 2017; 8(1): 262.
[http://dx.doi.org/10.1186/s13287-017-0710-3] [PMID: 29141662]
[78]
Mert T, Kurt AH, Altun İ, Celik A, Baran F, Gunay I. Pulsed magnetic field enhances therapeutic efficiency of mesenchymal stem cells in chronic neuropathic pain model. Bioelectromagnetics 2017; 38(4): 255-64.
[http://dx.doi.org/10.1002/bem.22038] [PMID: 28130880]
[79]
Xie J, Xiao D, Xu Y, et al. Up-regulation of immunomodulatory effects of mouse bone-marrow derived mesenchymal stem cells by tetrahydrocannabinol pre-treatment involving cannabinoid receptor CB2. Oncotarget 2016; 7(6): 6436-47.
[http://dx.doi.org/10.18632/oncotarget.7042] [PMID: 26824325]
[80]
Nalli Y, Dar MS, Bano N, et al. Analyzing the role of cannabinoids as modulators of Wnt/β-catenin signaling pathway for their use in the management of neuropathic pain. Bioorg Med Chem Lett 2019; 29(9): 1043-6.
[http://dx.doi.org/10.1016/j.bmcl.2019.03.013] [PMID: 30871771]
[81]
Vučković S, Srebro D, Vujović KS, Vučetić Č, Prostran M. Cannabinoids and Pain: New Insights From Old Molecules. Front Pharmacol 2018; 9: 1259.
[http://dx.doi.org/10.3389/fphar.2018.01259] [PMID: 30542280]
[82]
Zhang M, Dong L, Zou H, et al. Effects of Cannabinoid Type 2 Receptor Agonist AM1241 on Morphine-Induced Antinociception, Acute and Chronic Tolerance, and Dependence in Mice. J Pain 2018; 19(10): 1113-29.
[http://dx.doi.org/10.1016/j.jpain.2018.04.009] [PMID: 29729431]
[83]
Shupak NM, Prato FS, Thomas AW. Human exposure to a specific pulsed magnetic field: effects on thermal sensory and pain thresholds. Neurosci Lett 2004; 363(2): 157-62.
[http://dx.doi.org/10.1016/j.neulet.2004.03.069] [PMID: 15172106]
[84]
Yousefi F, Lavi Arab F, Saeidi K, Amiri H, Mahmoudi M. Various strategies to improve efficacy of stem cell transplantation in multiple sclerosis: Focus on mesenchymal stem cells and neuroprotection. J Neuroimmunol 2019; 328: 20-34.
[http://dx.doi.org/10.1016/j.jneuroim.2018.11.015] [PMID: 30557687]
[85]
Lu Y, Zhou Y, Zhang R, et al. Bone mesenchymal stem cell-derived extracellular vesicles promote recovery following spinal cord injury via improvement of the integrity of the blood-spinal cord barrier. Front Neurosci 2019; 13: 209.
[http://dx.doi.org/10.3389/fnins.2019.00209] [PMID: 30914918]
[86]
Ma Y, Dong L, Zhou D, et al. Extracellular vesicles from human umbilical cord mesenchymal stem cells improve nerve regeneration after sciatic nerve transection in rats. J Cell Mol Med 2019; 23(4): 2822-35.
[http://dx.doi.org/10.1111/jcmm.14190] [PMID: 30772948]
[87]
Zhao T, Sun F, Liu J, et al. Emerging role of mesenchymal stem cell-derived exosomes in regenerative medicine. Curr Stem Cell Res Ther 2019.
[http://dx.doi.org/10.2174/1574888X14666190228103230] [PMID: 30819086]
[88]
Ren K. Exosomes in perspective: a potential surrogate for stem cell therapy. Odontology 2018.
[PMID: 30324571]
[89]
Tracy SA, Ahmed A, Tigges JC, et al. A comparison of clinically relevant sources of mesenchymal stem cell-derived exosomes: Bone marrow and amniotic fluid. J Pediatr Surg 2019; 54(1): 86-90.
[http://dx.doi.org/10.1016/j.jpedsurg.2018.10.020] [PMID: 30361074]
[90]
Andriolo G, Provasi E, Lo Cicero V, et al. Exosomes from human cardiac progenitor cells for therapeutic applications: Development of a GMP-grade manufacturing method. Front Physiol 2018; 9: 1169.
[http://dx.doi.org/10.3389/fphys.2018.01169] [PMID: 30197601]
[91]
Tamkovich S, Tutanov O, Efimenko A, et al. Blood circulating exosomes contain distinguishable fractions of free and cell-surface-associated vesicles. Curr Mol Med 2019; 19(4): 273-85.
[http://dx.doi.org/10.2174/1566524019666190314120532] [PMID: 30868953]
[92]
Rosas-Hernandez H, Cuevas E, Raymick JB, et al. Characterization of serum exosomes from a transgenic mouse model of alzheimer’s disease. Curr Alzheimer Res 2019; 16(5): 388-95.
[http://dx.doi.org/10.2174/1567205016666190321155422] [PMID: 30907317]
[93]
Kumeda N, Ogawa Y, Akimoto Y, Kawakami H, Tsujimoto M, Yanoshita R. Characterization of membrane integrity and morphological stability of human salivary exosomes. Biol Pharm Bull 2017; 40(8): 1183-91.
[http://dx.doi.org/10.1248/bpb.b16-00891] [PMID: 28768999]
[94]
Katsiougiannis S, Chia D, Kim Y, Singh RP, Wong DT. Saliva exosomes from pancreatic tumor-bearing mice modulate NK cell phenotype and antitumor cytotoxicity. FASEB J 2017; 31(3): 998-1010.
[http://dx.doi.org/10.1096/fj.201600984R] [PMID: 27895106]
[95]
Machida T, Tomofuji T, Ekuni D, et al. MicroRNAs in salivary exosome as potential biomarkers of aging. Int J Mol Sci 2015; 16(9): 21294-309.
[http://dx.doi.org/10.3390/ijms160921294] [PMID: 26370963]
[96]
Mizutani K, Kawakami K, Horie K, et al. Urinary exosome as a potential biomarker for urinary tract infection. Cell Microbiol 2019; 21(7)e13020
[http://dx.doi.org/10.1111/cmi.13020] [PMID: 30817089]
[97]
Yazarlou F, Mowla SJ, Oskooei VK, et al. Urine exosome gene expression of cancer-testis antigens for prediction of bladder carcinoma. Cancer Manag Res 2018; 10: 5373-81.
[http://dx.doi.org/10.2147/CMAR.S180389] [PMID: 30464633]
[98]
Vall-Palomar M, Arévalo J, Ariceta G, Meseguer A. Establishment of urinary exosome-like vesicles isolation protocol for FHHNC patients and evaluation of different exosomal RNA extraction methods. J Transl Med 2018; 16(1): 278.
[http://dx.doi.org/10.1186/s12967-018-1651-z] [PMID: 30305086]
[99]
Guha D, Mukerji SS, Chettimada S, et al. Cerebrospinal fluid extracellular vesicles and neurofilament light protein as biomarkers of central nervous system injury in HIV-infected patients on antiretroviral therapy. AIDS 2019; 33(4): 615-25.
[http://dx.doi.org/10.1097/QAD.0000000000002121] [PMID: 30557159]
[100]
Manek R, Moghieb A, Yang Z, et al. Protein biomarkers and neuroproteomics characterization of microvesicles/exosomes from human cerebrospinal fluid following traumatic brain injury. Mol Neurobiol 2018; 55(7): 6112-28.
[http://dx.doi.org/10.1007/s12035-017-0821-y] [PMID: 29188495]
[101]
Kong FL, Wang XP, Li YN, Wang HX. The role of exosomes derived from cerebrospinal fluid of spinal cord injury in neuron proliferation in vitro. Artif Cells Nanomed Biotechnol 2018; 46(1): 200-5.
[http://dx.doi.org/10.1080/21691401.2017.1304408] [PMID: 28346015]
[102]
Li B, Hock A, Wu RY, et al. Bovine milk-derived exosomes enhance goblet cell activity and prevent the development of experimental necrotizing enterocolitis. PLoS One 2019; 14(1)e0211431
[http://dx.doi.org/10.1371/journal.pone.0211431] [PMID: 30699187]
[103]
Hock A, Miyake H, Li B, et al. Breast milk-derived exosomes promote intestinal epithelial cell growth. J Pediatr Surg 2017; 52(5): 755-9.
[http://dx.doi.org/10.1016/j.jpedsurg.2017.01.032] [PMID: 28188035]
[104]
Qin W, Tsukasaki Y, Dasgupta S, Mukhopadhyay N, Ikebe M, Sauter ER. Exosomes in human breast milk promote EMT. Clin Cancer Res 2016; 22(17): 4517-24.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-0135] [PMID: 27060153]
[105]
Baglio SR, Rooijers K, Koppers-Lalic D, et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res Ther 2015; 6: 127.
[http://dx.doi.org/10.1186/s13287-015-0116-z] [PMID: 26129847]
[106]
Li Z, Liu F, He X, Yang X, Shan F, Feng J. Exosomes derived from mesenchymal stem cells attenuate inflammation and demyelination of the central nervous system in EAE rats by regulating the polarization of microglia. Int Immunopharmacol 2019; 67: 268-80.
[http://dx.doi.org/10.1016/j.intimp.2018.12.001] [PMID: 30572251]
[107]
Chew JRJ, Chuah SJ, Teo KYW, et al. Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration. Acta Biomater 2019; 89: 252-64.
[http://dx.doi.org/10.1016/j.actbio.2019.03.021] [PMID: 30878447]
[108]
Goetzl EJ, Kapogiannis D, Schwartz JB, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J 2016; 30(12): 4141-8.
[http://dx.doi.org/10.1096/fj.201600816R] [PMID: 27601437]
[109]
Furuta T, Miyaki S, Ishitobi H, et al. Mesenchymal stem cell-derived exosomes promote fracture healing in a mouse model. Stem Cells Transl Med 2016; 5(12): 1620-30.
[http://dx.doi.org/10.5966/sctm.2015-0285] [PMID: 27460850]
[110]
Thomi G, Surbek D, Haesler V, Joerger-Messerli M, Schoeberlein A. Exosomes derived from umbilical cord mesenchymal stem cells reduce microglia-mediated neuroinflammation in perinatal brain injury. Stem Cell Res Ther 2019; 10(1): 105.
[http://dx.doi.org/10.1186/s13287-019-1207-z] [PMID: 30898154]
[111]
Mao F, Wu Y, Tang X, et al. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Relieve Inflammatory Bowel Disease in Mice. BioMed Res Int 2017.20175356760
[http://dx.doi.org/10.1155/2017/5356760] [PMID: 28589143]
[112]
Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab 2013; 33(11): 1711-5.
[http://dx.doi.org/10.1038/jcbfm.2013.152] [PMID: 23963371]
[113]
Fang Y, Zhang Y, Zhou J, Cao K. Adipose-derived mesenchymal stem cell exosomes: a novel pathway for tissues repair. Cell Tissue Bank 2019; 20(2): 153-61.
[http://dx.doi.org/10.1007/s10561-019-09761-y] [PMID: 30852701]
[114]
Wang Y, Yu D, Liu Z, et al. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther 2017; 8(1): 189.
[http://dx.doi.org/10.1186/s13287-017-0632-0] [PMID: 28807034]
[115]
Wu J, Kuang L, Chen C, et al. miR-100-5p-abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials 2019; 206: 87-100.
[http://dx.doi.org/10.1016/j.biomaterials.2019.03.022] [PMID: 30927715]
[116]
Xu G, Ao R, Zhi Z, Jia J, Yu B. miR-21 and miR-19b delivered by hMSC-derived EVs regulate the apoptosis and differentiation of neurons in patients with spinal cord injury. J Cell Physiol 2019; 234(7): 10205-17.
[http://dx.doi.org/10.1002/jcp.27690] [PMID: 30387159]

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