360
Views
7
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

The Therapeutic Effect of iMSC-Derived Small Extracellular Vesicles on Tendinopathy Related Pain Through Alleviating Inflammation: An in vivo and in vitro Study

ORCID Icon, , , , , , & show all
Pages 1421-1436 | Published online: 01 Mar 2022

References

  • Riel H, Lindstrøm C, Rathleff M, Jensen M, Olesen J. Prevalence and incidence rate of lower-extremity tendinopathies in a Danish general practice: a registry-based study. BMC Musculoskelet Disord. 2019;20(1):239. doi:10.1186/s12891-019-2629-6
  • De Vries A, Koolhaas W, Zwerver J, et al. The impact of patellar tendinopathy on sports and work performance in active athletes. Res Sports Med. 2017;25(3):253–265. doi:10.1080/15438627.2017.1314292
  • Chianca V, Albano D, Messina C, et al. Rotator cuff calcific tendinopathy: from diagnosis to treatment. Acta Biomed. 2018;89(1–s):186–196. doi:10.23750/abm.v89i1-S.7022
  • Silbernagel KG, Hanlon S, Sprague A. Current clinical concepts: conservative management of achilles tendinopathy. J Athl Train. 2020;55(5):438–447. doi:10.4085/1062-6050-356-19
  • Choi HJ, Choi S, Kim JG, et al. Enhanced tendon restoration effects of anti-inflammatory, lactoferrin-immobilized, heparin-polymeric nanoparticles in an Achilles tendinitis rat model. Carbohydr Polym. 2020;241:116284. doi:10.1016/j.carbpol.2020.116284
  • Kang S, Yoon JS, Lee JY, Kim HJ, Park K, Kim SE. Long-term local PDGF delivery using porous microspheres modified with heparin for tendon healing of rotator cuff tendinitis in a rabbit model. Carbohydr Polym. 2019;209:372–381. doi:10.1016/j.carbpol.2019.01.017
  • Kim SE, Yun Y-P, Shim K-S, Jeon DI, Park K, Kim H-J. In vitro and in vivo anti-inflammatory and tendon-healing effects in Achilles tendinopathy of long-term curcumin delivery using porous microspheres. J Ind Eng Chem. 2018;58:123–130. doi:10.1016/j.jiec.2017.09.016
  • Abate M, Silbernagel K, Siljeholm C, et al. Pathogenesis of tendinopathies: inflammation or degeneration? Arthritis Res Ther. 2009;11(3):235. doi:10.1186/ar2723
  • Cazzola M, Atzeni F, Boccassini L, Cassisi G, Sarzi-Puttini P. Physiopathology of pain in rheumatology. Reumatismo. 2014;66(1):4–13. doi:10.4081/reumatismo.2014.758
  • Tang C, Chen Y, Huang J, et al. The roles of inflammatory mediators and immunocytes in tendinopathy. J Orthop Translat. 2018;14:23–33. doi:10.1016/j.jot.2018.03.003
  • Dean BJ, Gettings P, Dakin SG, Carr AJ. Are inflammatory cells increased in painful human tendinopathy? A systematic review. Br J Sports Med. 2016;50(4):216–220. doi:10.1136/bjsports-2015-094754
  • Schubert TE, Weidler C, Lerch K, Hofstädter F, Straub RH. Achilles tendinosis is associated with sprouting of substance P positive nerve fibres. Ann Rheum Dis. 2005;64(7):1083–1086. doi:10.1136/ard.2004.029876
  • Millar NL, Hueber AJ, Reilly JH, et al. Inflammation is present in early human tendinopathy. Am J Sports Med. 2010;38(10):2085–2091. doi:10.1177/0363546510372613
  • Manning CN, Martel C, Sakiyama-Elbert SE, et al. Adipose-derived mesenchymal stromal cells modulate tendon fibroblast responses to macrophage-induced inflammation in vitro. Stem Cell Res Ther. 2015;6(1):74. doi:10.1186/s13287-015-0059-4
  • Qi J, Chi L, Maloney M, Yang X, Bynum D, Banes A. Interleukin-1beta increases elasticity of human bioartificial tendons. Tissue Eng. 2006;12(10):2913–2925. doi:10.1089/ten.2006.12.2913
  • Rees J, Stride M, Scott A. Tendons–time to revisit inflammation. Br J Sports Med. 2014;48(21):1553–1557. doi:10.1136/bjsports-2012-091957
  • Spang C, Renström L, Alfredson H, Forsgren S. Marked expression of TNF receptors in human peritendinous tissues including in nerve fascicles with axonal damage - Studies on tendinopathy and tennis elbow. J Musculoskelet Neuronal Interact. 2017;17(3):226–236.
  • Takano S, Uchida K, Miyagi M, et al. Nerve growth factor regulation by TNF-α and IL-1β in synovial macrophages and fibroblasts in osteoarthritic mice. J Immunol Res. 2016;2016:5706359. doi:10.1155/2016/5706359
  • Nagura N, Kenmoku T, Uchida K, Nakawaki M, Inoue G, Takaso M. Nerve growth factor continuously elevates in a rat rotator cuff tear model. J Shoulder Elbow Surg. 2019;28(1):143–148. doi:10.1016/j.jse.2018.06.030
  • Brown M, Murphy F, Radin D, Davignon I, Smith M, West C. Tanezumab reduces osteoarthritic knee pain: results of a randomized, double-blind, placebo-controlled Phase III trial. J Pain. 2012;13(8):790–798. doi:10.1016/j.jpain.2012.05.006
  • Markman J, Bolash R, McAlindon T, et al. Tanezumab for chronic low back pain: a randomized, double-blind, placebo- and active-controlled, Phase 3 study of efficacy and safety. Pain. 2020;161(9):2068–2078. doi:10.1097/j.pain.0000000000001928
  • Munir H, Ward L, Mcgettrick HM. Mesenchymal stem cells as endogenous regulators of inflammation. Stromal Immunol. 2018;4:73–98.
  • Rasmusson I. Immune modulation by mesenchymal stem cells. Exp Cell Res. 2006;312(12):2169–2179. doi:10.1016/j.yexcr.2006.03.019
  • Hu GW, Li Q, Niu X, et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res Ther. 2015;6:1–5.
  • Van Niel G, d’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–218.
  • Yang Y, Zhu Z, Gao R, et al. Controlled release of MSC-derived small extracellular vesicles by an injectable Diels-Alder crosslinked hyaluronic acid/PEG hydrogel for osteoarthritis improvement. Acta Biomater. 2021;128:163–174.
  • Zhu Y, Wang Y, Zhao B, et al. Comparison of exosomes secreted by induced pluripotent stem cell-derived mesenchymal stem cells and synovial membrane-derived mesenchymal stem cells for the treatment of osteoarthritis. Stem Cell Res Ther. 2017;8(1):64. doi:10.1186/s13287-017-0510-9
  • Shen H, Yoneda S, Abu-Amer Y, Guilak F, Gelberman RH. Stem cell-derived extracellular vesicles attenuate the early inflammatory response after tendon injury and repair. J Orthop Res. 2020;38(1):117–127. doi:10.1002/jor.24406
  • Yu H, Cheng J, Shi W, et al. Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells. Acta Biomater. 2020;106:328–341. doi:10.1016/j.actbio.2020.01.051
  • Xia Y, Ling X, Hu G, et al. Small extracellular vesicles secreted by human iPSC-derived MSC enhance angiogenesis through inhibiting STAT3-dependent autophagy in ischemic stroke. Stem Cell Res Ther. 2020;11(1):313. doi:10.1186/s13287-020-01834-0
  • Chen B, Sun Y, Zhang J, et al. Human embryonic stem cell-derived exosomes promote pressure ulcer healing in aged mice by rejuvenating senescent endothelial cells. Stem Cell Res Ther. 2019;10(1):142. doi:10.1186/s13287-019-1253-6
  • Tian Y, Ma L, Gong M, et al. Protein profiling and sizing of extracellular vesicles from colorectal cancer patients via flow cytometry. ACS Nano. 2018;12(1):671–680. doi:10.1021/acsnano.7b07782
  • Eskildsen S, Berkoff D, Kallianos S, Weinhold P. The use of an IL1-receptor antagonist to reverse the changes associated with established tendinopathy in a rat model. Scand J Med Sci Sports. 2019;29(1):82–88. doi:10.1111/sms.13310
  • Da Ré Guerra F, Vieira C, Marques P, Oliveira L, Pimentel E. Low level laser therapy accelerates the extracellular matrix reorganization of inflamed tendon. Tissue Cell. 2017;49(4):483–488. doi:10.1016/j.tice.2017.05.006
  • Mogil J. Animal models of pain: progress and challenges. Nat Rev Neurosci. 2009;10(4):283–294. doi:10.1038/nrn2606
  • Otis C, Gervais J, Guillot M, et al. Concurrent validity of different functional and neuroproteomic pain assessment methods in the rat osteoarthritis monosodium iodoacetate (MIA) model. Arthritis Res Ther. 2016;18:150. doi:10.1186/s13075-016-1047-5
  • Malfait A, Little C, McDougall J. A commentary on modelling osteoarthritis pain in small animals. Osteoarthritis Cartilage. 2013;21(9):1316–1326. doi:10.1016/j.joca.2013.06.003
  • Cong GT, Lebaschi AH, Camp CL, et al. Evaluating the role of subacromial impingement in rotator cuff tendinopathy: development and analysis of a novel murine model. J Orthop Res. 2018;36(10):2780–2788. doi:10.1002/jor.24026
  • Jensen EC. Quantitative analysis of histological staining and fluorescence using ImageJ. Anat Rec. 2013;296(3):378–381. doi:10.1002/ar.22641
  • Xiao M, Leonardi EA, Sharpe O, et al. Soaking of autologous tendon grafts in vancomycin before implantation does not lead to tenocyte cytotoxicity. Am J Sports Med. 2020;48(12):3081–3086. doi:10.1177/0363546520951815
  • Wiklander OP, Nordin JZ, O’Loughlin A, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316. doi:10.3402/jev.v4.26316
  • Tsuzaki M, Guyton G, Garrett W, et al. IL-1 beta induces COX2, MMP-1, −3 and −13, ADAMTS-4, IL-1 beta and IL-6 in human tendon cells. J Orthop Res. 2003;21(2):256–264. doi:10.1016/S0736-0266(02)00141-9
  • Maeda E, Kuroyanagi K, Ando Y, Matsumoto T. Effects of substrate stiffness on morphology and MMP-1 gene expression in tenocytes stimulated with Interleukin-1β. J Orthop Res. 2020;38(1):150–159. doi:10.1002/jor.24403
  • Piel MJ, Kroin JS, van Wijnen AJ, Kc R, Im H-J. Pain assessment in animal models of osteoarthritis. Gene. 2014;537(2):184–188. doi:10.1016/j.gene.2013.11.091
  • Yang M, Li CJ, Sun X, et al. MiR-497~195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch and HIF-1α activity. Nat Commun. 2017;8:16003. doi:10.1038/ncomms16003
  • Wang C, Zhang Y, Zhang G, Yu W, He Y. Adipose stem cell-derived exosomes ameliorate chronic rotator cuff tendinopathy by regulating macrophage polarization: from a mouse model to a study in human tissue. Am J Sports Med. 2021;49(9):2321–2331. doi:10.1177/03635465211020010
  • Smith B, Selfe J, Thacker D, et al. Incidence and prevalence of patellofemoral pain: a systematic review and meta-analysis. PLoS One. 2018;13(1):e0190892. doi:10.1371/journal.pone.0190892
  • Everhart JS, Cole D, Sojka JH, et al. Treatment options for patellar tendinopathy: a systematic review. Arthroscopy. 2017;33(4):861–872. doi:10.1016/j.arthro.2016.11.007
  • Halpern AA, Horowitz BG, Nagel DA. Tendon ruptures associated with corticosteroid therapy. West J Med. 1977;127(5):378–382.
  • Lebaschi A, Deng X, Zong J, et al. Animal models for rotator cuff repair. Ann N Y Acad Sci. 2016;1383(1):43–57. doi:10.1111/nyas.13203
  • Berberich P, Hoheisel U, Mense S. Effects of a carrageenan-induced myositis on the discharge properties of group III and IV muscle receptors in the cat. J Neurophysiol. 1988;59(5):1395–1409. doi:10.1152/jn.1988.59.5.1395
  • Radhakrishnan R, Moore S, Sluka K. Unilateral carrageenan injection into muscle or joint induces chronic bilateral hyperalgesia in rats. Pain. 2003;104(3):567–577. doi:10.1016/S0304-3959(03)00114-3
  • Rivot J, Montagne-Clavel J, Besson J. Subcutaneous formalin and intraplantar carrageenan increase nitric oxide release as measured by in vivo voltammetry in the spinal cord. Eur j Pain. 2002;6(1):25–34. doi:10.1053/eujp.2001.0268
  • Vieira C, De Aro A, Da Ré Guerra F, De Oliveira L, De Almeida MS, Pimentel E. Inflammatory process induced by carrageenan in adjacent tissue triggers the acute inflammation in deep digital flexor tendon of rats. Anat Rec. 2013;296(8):1187–1195. doi:10.1002/ar.22729
  • Çınar B, Çirci E, Balçık C, Güven G, Akpınar S, Derincek A. The effects of extracorporeal shock waves on carrageenan-induced Achilles tendinitis in rats: a biomechanical and histological analysis. Acta Orthop Traumatol Turc. 2013;47(4):266–272. doi:10.3944/AOTT.2013.2784
  • 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):215–224. doi:10.1002/sctm.18-0053
  • Di Cesare Mannelli L, Tenci B, Micheli L. Adipose-derived stem cells decrease pain in a rat model of oxaliplatin-induced neuropathy: role of VEGF-A modulation. Neuropharmacology. 2018;131:166–175. doi:10.1016/j.neuropharm.2017.12.020
  • Siniscalco D, Giordano C, Galderisi U. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci. 2011;5:79. doi:10.3389/fnint.2011.00079
  • Zhang S, Teo KYW, Chuah SJ, Lai RC, Lim SK, Toh WS. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials. 2019;200:35–47. doi:10.1016/j.biomaterials.2019.02.006
  • Peng X, Guo H, Yuan J, et al. Extracellular vesicles released from hiPSC-derived MSCs attenuate chronic prostatitis/chronic pelvic pain syndrome in rats by immunoregulation. Stem Cell Res Ther. 2021;12(1):198. doi:10.1186/s13287-021-02269-x
  • Wang Y, He G, Tang H, et al. Aspirin inhibits inflammation and scar formation in the injury tendon healing through regulating JNK/STAT-3 signalling pathway. Cell Prolif. 2019;52(4):e12650. doi:10.1111/cpr.12650
  • Surbek M, Tse W, Moriggl R, Han X. A centric view of JAK/STAT5 in intestinal homeostasis, infection, and inflammation. Cytokine. 2021;139:155392. doi:10.1016/j.cyto.2020.155392
  • Kato A. Group 2 innate lymphoid cells in airway diseases. Chest. 2019;156(1):141–149. doi:10.1016/j.chest.2019.04.101
  • Xie B, Wang S, Jiang N, Li JJ. Cyclin B1/CDK1-regulated mitochondrial bioenergetics in cell cycle progression and tumor resistance. Cancer Lett. 2019;443:56–66. doi:10.1016/j.canlet.2018.11.019
  • Fry AM, O’Regan L, Sabir SR, Bayliss R. Cell cycle regulation by the NEK family of protein kinases. J Cell Sci. 2012;125(Pt 19):4423–4433. doi:10.1242/jcs.111195
  • Horning AM, Wang Y, Lin CK, et al. Single-cell RNA-seq reveals a subpopulation of prostate cancer cells with enhanced cell-cycle-related transcription and attenuated androgen response. Cancer Res. 2018;78(4):853–864. doi:10.1158/0008-5472.CAN-17-1924
  • Van Doren SR. Matrix metalloproteinase interactions with collagen and elastin. Matrix Biol. 2015;44–46:224–231. doi:10.1016/j.matbio.2015.01.005
  • Panwar P, Butler GS, Jamroz A, Azizi P, Overall CM, Brömme D. Aging-associated modifications of collagen affect its degradation by matrix metalloproteinases. Matrix Biol. 2018;65:30–44. doi:10.1016/j.matbio.2017.06.004