Pulsed electromagnetic fields potentiate the paracrine function of mesenchymal stem cells for cartilage regeneration

Dinesh Parate, Nurul Dinah Kadir, Cenk Celik, Eng Hin Lee, James H P Hui, Alfredo Franco-Obregón, Zheng Yang, Dinesh Parate, Nurul Dinah Kadir, Cenk Celik, Eng Hin Lee, James H P Hui, Alfredo Franco-Obregón, Zheng Yang

Abstract

Background: The mesenchymal stem cell (MSC) secretome, via the combined actions of its plethora of biologically active factors, is capable of orchestrating the regenerative responses of numerous tissues by both eliciting and amplifying biological responses within recipient cells. MSCs are "environmentally responsive" to local micro-environmental cues and biophysical perturbations, influencing their differentiation as well as secretion of bioactive factors. We have previously shown that exposures of MSCs to pulsed electromagnetic fields (PEMFs) enhanced MSC chondrogenesis. Here, we investigate the influence of PEMF exposure over the paracrine activity of MSCs and its significance to cartilage regeneration.

Methods: Conditioned medium (CM) was generated from MSCs subjected to either 3D or 2D culturing platforms, with or without PEMF exposure. The paracrine effects of CM over chondrocytes and MSC chondrogenesis, migration and proliferation, as well as the inflammatory status and induced apoptosis in chondrocytes and MSCs was assessed.

Results: We show that benefits of magnetic field stimulation over MSC-derived chondrogenesis can be partly ascribed to its ability to modulate the MSC secretome. MSCs cultured on either 2D or 3D platforms displayed distinct magnetic sensitivities, whereby MSCs grown in 2D or 3D platforms responded most favorably to PEMF exposure at 2 mT and 3 mT amplitudes, respectively. Ten minutes of PEMF exposure was sufficient to substantially augment the chondrogenic potential of MSC-derived CM generated from either platform. Furthermore, PEMF-induced CM was capable of enhancing the migration of chondrocytes and MSCs as well as mitigating cellular inflammation and apoptosis.

Conclusions: The findings reported here demonstrate that PEMF stimulation is capable of modulating the paracrine function of MSCs for the enhancement and re-establishment of cartilage regeneration in states of cellular stress. The PEMF-induced modulation of the MSC-derived paracrine function for directed biological responses in recipient cells or tissues has broad clinical and practical ramifications with high translational value across numerous clinical applications.

Keywords: Cartilage; Mesenchymal stem cells; Paracrine; Pulse electromagnetic fields.

Conflict of interest statement

AF-O is an inventor on patent WO 2019/17863 A1, System and Method for Applying Pulsed Electromagnetic Fields, and contributes to QuantumTx Pte. Ltd., which elaborates on the use of similar magnetic fields for human use. All other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Schematic illustration of the generation and functional analysis of the CM from MSCs subjected to either 3D or 2D culturing platforms, with (PCM) or without (CCM) PEMF exposure
Fig. 2
Fig. 2
Brief PEMF exposure stimulates MSC-derived paracrine activity to promote chondrogenesis. MSCs in 3D pellet cultures undergoing chondrogenesis were subjected to PEMFs at either 0 or 2 mT for 10 min. The conditioned media generated after 24 h of PEMF exposure (PCM) was either replaced with age-matched media from unexposed sister cultures (CM-dep; CM-deprived), or transferred to age-matched exposed or unexposed sister cultures. Real-time PCR analysis of cartilaginous markers were performed after 7 days of differentiation and normalized to GAPDH. Results were presented as fold-changes relative to the level in undifferentiated MSCs. Data shown are means ± SD, n = 6 from 2 independent experiments. * denotes significance differences compared to non-PEMFed controls (red bars); # denotes significance differences compared to CM-deprived (CM-dep) condition; @ denotes significance differences compared to treatment with PCM alone
Fig. 3
Fig. 3
Effect of PEMF-induced conditioned media (CM) from 3D cultured MSCs on chondrogenic differentiation of MSCs. MSCs in 3D culture were subjected to PEMF exposure at different intensities and durations. The generated CM was collected 24 h post-PEMF exposure and tested for chondrogenic effect over naive (unexposed) MSC pellet cultures undergoing chondrogenic differentiation in the absence or presence of TGFβ. a Real-time PCR analysis of cartilaginous and hypertrophic marker expression after 7 days of differentiation normalized to GAPDH and presented as fold-changes relative to levels in undifferentiated MSCs. The expression of hypertrophic markers was presented as the ratio to Col 2 expression. b Quantification of cartilaginous extracellular matrix macromolecules generated by the differentiated MSCs (+TGFβ) after 21 days of differentiation. Data shown represent means ± SD, n = 6 from 2 independent experiments. * denotes significance differences compared to non-PEMFed (CCM, 0 mT) controls (red dash lines or red bars). # denotes significance differences compared to PEMFed CM (PCM) generated at 2 mT, 10 min
Fig. 4
Fig. 4
Effects of PEMF-induced conditioned media (CM) harvested from 2D cultures of MSCs over chondrogenic differentiation of MSCs. CM was collected 24 h post-PEMF exposure and tested for chondrogenic effect over naive (unexposed) MSC pellet cultures undergoing chondrogenic differentiation in the presence of TGFβ. a Real-time PCR analysis of cartilaginous and hypertrophic marker expression after 7 days of differentiation normalized to GAPDH and presented as fold-change relative to levels in undifferentiated MSCs. Expression of hypertrophic markers was presented as ratio to Col 2 expression. b Quantification of cartilaginous extracellular matrix macromolecules generated by differentiated MSCs after 21 days of differentiation. Data shown are means ± SD, n = 6 from 2 independent experiments. * denotes significance differences compare to non-PEMFed CM (0 mT) controls (red dash lines). # denotes significance differences compared to PEMFed CM (PCM) generated at 3 mT, 10 min exposure
Fig. 5
Fig. 5
Effects of PEMF-induced conditioned medium (CM) on chondrocyte redifferentiation. CM was generated from MSCs cultured on 2D and 3D platforms with and without PEMF exposure at 3 mT and 2 mT, respectively, or unexposed. CM collected 24 h post-PEMF exposure was administered to chondrocyte pellet cultures. a Real-time PCR analysis of cartilaginous marker expression after 7 days of redifferentiation was normalized to GAPDH and presented as a fold-change relative to the level expressed in day 0 chondrocytes. Col 1 and Col 10 expressions are shown as ratios relative to Col 2 expression. b Quantification of cartilaginous extracellular matrix macromolecules generated by chondrocytes after 21 days of redifferentiation. Data shown represent means ± SD, n = 6 from 2 independent experiments. * denotes significant increases compare to respective non-PEMFed CM (CCM, 0 mT) (red solid bars)
Fig. 6
Fig. 6
Effect of PEMF-induced conditioned medium (CM) on chondrocyte and MSC migration and proliferation. CM was derived from 2D cultured MSCs in response to 0 mT (CCM) or 3 mT (PCM) PEMF exposure. a Migration of chondrocytes or MSCs was analyzed using a transwell culture. Migrated cells were assessed by measuring the number of cells on the underside of the transwell filter after H&E staining. b Cell proliferation was determined by Picogreen DNA Assay. Data shown represent means ± SD, n = 6 from 2 independent experiments. * denotes significant difference compared to the negative control (Expansion media + 0.5% FBS); # denotes significant differences compared to positive controls (Expansion media + 10% FBS); @ denotes significant differences compared to the non-PEMFed CCM
Fig. 7
Fig. 7
Effect of PEMF-induced conditioned medium (CM) on inflamed chondrocytes. Inflammation was induced in chondrocytes with 5 ng/ml IL-1β for 24 h. CCM or PCM was administered to chondrocytes 24 h post inflammation induction. Real-time PCR analysis of cartilaginous and inflammatory markers and NOS activity were performed at 24 h (plain bars) and 48 h (hatched bars) post-supplementation with CM and normalized to GAPDH, presented as a fold-change relative to the level in non-treated (day 0) chondrocytes. Data shown represent means ± SD, n = 6 from 2 independent experiments. * denotes significant differences compared to the non-inflamed controls (no IL-1β treatment); # denotes significant differences compared to the inflammation controls (IL-1β alone treatment) and @ denotes significant differences compared to respective CCM. Col 2 = type II collagen; COX-2 = cycloxigenase-2; IL-1β = interleukin-1β; IL-6 = interleukin-6; MMP-13 = metalloproteinase 13; NOS = nitric oxide synthase
Fig. 8
Fig. 8
Effect of PEMF-induced conditioned medium (CM) on inflamed MSCs pre- (a) and post- (b) chondrogenic induction. a Inflammation was induced in MSCs with 5 ng/ml IL-1β for 24 h before administration of CCM or PCM generated from 2D MSC cultures. Expression of inflammatory genes and NOS activity was assayed at 24 (plain bars) and 48 h (hatched bars) after CM treatment. b MSCs undergoing chondrogenesis in 3D pellet cultures were treated with 5 ng/ml IL-1β 24 h prior to the administration of CCM or PCM. Expression cartilaginous markers was analyzed after 7 days of differentiation, normalized to GAPDH and presented as fold-changes relative to levels in undifferentiated MSCs. Data shown represent means ± SD, n = 6 from 2 independent experiments. * denotes significant differences compared to the non-inflamed controls (no IL-1β treatment), # denotes significant differences compared to the inflammation controls (IL-1β alone treatment), and @ denotes significant differences compared to respective CCM. Col 2 = type II collagen; COX-2 = cycloxigenase-2; IL-1β = interleukin-1β; IL-6 = interleukin-6; MMP-13 = metalloproteinase 13; NOS = nitric oxide synthase; Sox9 = SRY-Box 9
Fig. 9
Fig. 9
Effect of PEMF-induced conditioned medium (CM) on the apoptotic status of MSCs and chondrocytes. Chondrocytes (a) or MSCs (b) were treated with Staurosporin (SPN; 200 nM) in conjunction with supplementation with either CCM or PCM for 2 h. Apoptotic activity was determined by Caspase 3/7 activity and was presented as relative fluorescence units (RFU). Data shown represent means ± SD, n = 6 from 2 independent experiments. * denote significant differences compared to no Staurosporin treatment; # denotes significant differences compared to Staurosporin treatment alone and @ denotes significant differences relative to CCM
Fig. 10
Fig. 10
PEMF exposure modulates the secretion of MSC-derived paracrine factors. a 2D cultures of MSCs without (0 mT) and with PEMF exposure at 3 mT for 10 min were subjected to real-time PCR analysis after 24 h. Data shown represents means ± SD, n = 6 from 3 independent experiments. b Heatmap generated from an antibody microarray performed on the CM showing differences in the secretion profile of paracrine factors from MSCs. BMP = bone morphogenetic protein; COX-2 = cycloxigenase-2; IGF = insulin-like growth factor; IL-1ra: interleukin 1 receptor antagonist; TGF = transforming growth factor; TSP = thrombospondin

References

    1. Marcacci M, Filardo G, Kon E. Treatment of cartilage lesions: what works and why? Injury. 2013;44(Suppl 1):S11–S15. doi: 10.1016/S0020-1383(13)70004-4.
    1. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum. 2012;64(6):1697–1707. doi: 10.1002/art.34453.
    1. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147. doi: 10.1126/science.284.5411.143.
    1. Toh WS, Brittberg M, Farr J, Foldager CB, Gomoll AH, Hui JH, et al. Cellular senescence in aging and osteoarthritis. Acta Orthop. 2016;87(sup363):6–14. doi: 10.1080/17453674.2016.1235087.
    1. Somoza RA, Welter JF, Correa D, Caplan AI. Chondrogenic differentiation of mesenchymal stem cells: challenges and unfulfilled expectations. Tissue Eng Part B Rev. 2014;20(6):596–608. doi: 10.1089/ten.teb.2013.0771.
    1. Wu L, Leijten JC, Georgi N, Post JN, van Blitterswijk CA, Karperien M. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng Part A. 2011;17(9–10):1425–1436. doi: 10.1089/ten.tea.2010.0517.
    1. Levorson EJ, Santoro M, Kasper FK, Mikos AG. Direct and indirect co-culture of chondrocytes and mesenchymal stem cells for the generation of polymer/extracellular matrix hybrid constructs. Acta Biomater. 2014;10(5):1824–1835. doi: 10.1016/j.actbio.2013.12.026.
    1. Maumus M, Jorgensen C, Noel D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. Biochimie. 2013;95(12):2229–2234. doi: 10.1016/j.biochi.2013.04.017.
    1. Ha CW, Park YB, Kim SH, Lee HJ. Intra-articular mesenchymal stem cells in osteoarthritis of the knee: a systematic review of clinical outcomes and evidence of cartilage repair. Arthroscopy. 2019;35(1):277–288. doi: 10.1016/j.arthro.2018.07.028.
    1. Kupcova SH. Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie. 2013;95(12):2196–2211. doi: 10.1016/j.biochi.2013.07.015.
    1. Van Linthout S, Stamm C, Schultheiss HP, Tschope C. Mesenchymal stem cells and inflammatory cardiomyopathy: cardiac homing and beyond. Cardiol Res Pract. 2011;2011:757154. doi: 10.4061/2011/757154.
    1. Oh JY, Lee RH, Yu JM, Ko JH, Lee HJ, Ko AY, et al. Intravenous mesenchymal stem cells prevented rejection of allogeneic corneal transplants by aborting the early inflammatory response. Mol Ther. 2012;20(11):2143–2152. doi: 10.1038/mt.2012.165.
    1. Zhang S, Chu WC, Lai RC, Lim SK, Hui JH, Toh WS. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthr Cartil. 2016;24(12):2135–2140. doi: 10.1016/j.joca.2016.06.022.
    1. Zhang S, Chuah SJ, Lai RC, Hui JHP, Lim SK, Toh WS. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials. 2018;156:16–27. doi: 10.1016/j.biomaterials.2017.11.028.
    1. Wang Y, Yu D, Liu Z, Zhou F, Dai J, Wu B, 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. doi: 10.1186/s13287-017-0632-0.
    1. Cosenza S, Ruiz M, Toupet K, Jorgensen C, Noel D. Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci Rep. 2017;7(1):16214. doi: 10.1038/s41598-017-15376-8.
    1. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54. doi: 10.1038/emm.2013.94.
    1. Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the microenvironment on mesenchymal stem cell paracrine signaling: opportunities to engineer the therapeutic effect. Stem Cells Dev. 2017;26(9):617–631. doi: 10.1089/scd.2016.0349.
    1. Rodriguez TM, Saldias A, Irigo M, Zamora JV, Perone MJ, Dewey RA. Effect of TGF-beta1 stimulation on the secretome of human adipose-derived Mesenchymal stromal cells. Stem Cells Transl Med. 2015;4(8):894–898. doi: 10.5966/sctm.2015-0012.
    1. Li C, Li G, Liu M, Zhou T, Zhou H. Paracrine effect of inflammatory cytokine-activated bone marrow mesenchymal stem cells and its role in osteoblast function. J Biosci Bioeng. 2016;121(2):213–219. doi: 10.1016/j.jbiosc.2015.05.017.
    1. Teixeira FG, Panchalingam KM, Anjo SI, Manadas B, Pereira R, Sousa N, et al. Do hypoxia/normoxia culturing conditions change the neuroregulatory profile of Wharton jelly mesenchymal stem cell secretome? Stem Cell Res Ther. 2015;6:133. doi: 10.1186/s13287-015-0124-z.
    1. Paquet J, Deschepper M, Moya A, Logeart-Avramoglou D, Boisson-Vidal C, Petite H. Oxygen tension regulates human mesenchymal stem cell paracrine functions. Stem Cells Transl Med. 2015;4(7):809–821. doi: 10.5966/sctm.2014-0180.
    1. Sullivan KE, Quinn KP, Tang KM, Georgakoudi I, Black LD., 3rd Extracellular matrix remodeling following myocardial infarction influences the therapeutic potential of mesenchymal stem cells. Stem Cell Res Ther. 2014;5(1):14. doi: 10.1186/scrt403.
    1. Jose S, Hughbanks ML, Binder BY, Ingavle GC, Leach JK. Enhanced trophic factor secretion by mesenchymal stem/stromal cells with glycine-histidine-lysine (GHK)-modified alginate hydrogels. Acta Biomater. 2014;10(5):1955–1964. doi: 10.1016/j.actbio.2014.01.020.
    1. Seib FP, Prewitz M, Werner C, Bornhauser M. Matrix elasticity regulates the secretory profile of human bone marrow-derived multipotent mesenchymal stromal cells (MSCs) Biochem Biophys Res Commun. 2009;389(4):663–667. doi: 10.1016/j.bbrc.2009.09.051.
    1. Yang H, Nguyen KT, Leong DT, Tan NS, Tay CY. Soft material approach to induce oxidative stress in mesenchymal stem cells for functional tissue repair. ACS Appl Mater Interfaces. 2016;8(40):26591–26599. doi: 10.1021/acsami.6b09222.
    1. Santos JM, Camoes SP, Filipe E, Cipriano M, Barcia RN, Filipe M, et al. Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing. Stem Cell Res Ther. 2015;6:90. doi: 10.1186/s13287-015-0082-5.
    1. Lee EJ, Park SJ, Kang SK, Kim GH, Kang HJ, Lee SW, et al. Spherical bullet formation via E-cadherin promotes therapeutic potency of mesenchymal stem cells derived from human umbilical cord blood for myocardial infarction. Mol Ther. 2012;20(7):1424–1433. doi: 10.1038/mt.2012.58.
    1. Bartosh TJ, Ylostalo JH, Mohammadipoor A, Bazhanov N, Coble K, Claypool K, et al. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc Natl Acad Sci U S A. 2010;107(31):13724–13729. doi: 10.1073/pnas.1008117107.
    1. Chatterjee S, Fisher AB. Mechanotransduction: forces, sensors, and redox signaling. Antioxid Redox Signal. 2014;20(6):868–871. doi: 10.1089/ars.2013.5753.
    1. Kasper G, Dankert N, Tuischer J, Hoeft M, Gaber T, Glaeser JD, et al. Mesenchymal stem cells regulate angiogenesis according to their mechanical environment. Stem Cells. 2007;25(4):903–910. doi: 10.1634/stemcells.2006-0432.
    1. Gardner OF, Fahy N, Alini M, Stoddart MJ. Differences in human mesenchymal stem cell secretomes during chondrogenic induction. Eur Cell Mater. 2016;31:221–235. doi: 10.22203/eCM.v031a15.
    1. Scheele C, Nielsen S, Pedersen BK. ROS and myokines promote muscle adaptation to exercise. Trends Endocrinol Metab. 2009;20(3):95–99. doi: 10.1016/j.tem.2008.12.002.
    1. Parate D, Franco-Obregon A, Frohlich J, Beyer C, Abbas AA, Kamarul T, et al. Enhancement of mesenchymal stem cell chondrogenesis with short-term low intensity pulsed electromagnetic fields. Sci Rep. 2017;7(1):9421. doi: 10.1038/s41598-017-09892-w.
    1. Yap JL, Tai YK, Fröhlich J, Fong CHH, Yin JN, Foo ZL, Ramanan S, Beyer C, Toh SJ, Casarosa M, Bharathy N, Kala MP, Egli M, Taneja, Lee CN, Franco-Obregón A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism. FASEB J. 2019;33(11):12853–12872. 10.1096/fj.201900057R.
    1. Aaron RK, Wang S, Ciombor DM. Upregulation of basal TGFbeta1 levels by EMF coincident with chondrogenesis--implications for skeletal repair and tissue engineering. J Orthop Res. 2002;20(2):233–240. doi: 10.1016/S0736-0266(01)00084-5.
    1. Jansen JH, van der Jagt OP, Punt BJ, Verhaar JA, van Leeuwen JP, Weinans H, et al. Stimulation of osteogenic differentiation in human osteoprogenitor cells by pulsed electromagnetic fields: an in vitro study. BMC Musculoskelet Disord. 2010;11:188. doi: 10.1186/1471-2474-11-188.
    1. Amin HD, Brady MA, St-Pierre JP, Stevens MM, Overby DR, Ethier CR. Stimulation of chondrogenic differentiation of adult human bone marrow-derived stromal cells by a moderate-strength static magnetic field. Tissue Eng Part A. 2014;20(11–12):1612–1620. doi: 10.1089/ten.tea.2013.0307.
    1. Kwon HJ, Lee GS, Chun H. Electrical stimulation drives chondrogenesis of mesenchymal stem cells in the absence of exogenous growth factors. Sci Rep. 2016;6:39302. doi: 10.1038/srep39302.
    1. Varani K, Padovan M, Vincenzi F, Targa M, Trotta F, Govoni M, et al. A2A and A3 adenosine receptor expression in rheumatoid arthritis: upregulation, inverse correlation with disease activity score and suppression of inflammatory cytokine and metalloproteinase release. Arthritis Res Ther. 2011;13(6):R197. doi: 10.1186/ar3527.
    1. Varani K, Vincenzi F, Ravani A, Pasquini S, Merighi S, Gessi S, et al. Adenosine receptors as a biological pathway for the anti-inflammatory and beneficial effects of low frequency low energy pulsed electromagnetic fields. Mediat Inflamm. 2017;2017:2740963. doi: 10.1155/2017/2740963.
    1. De Mattei M, Varani K, Masieri FF, Pellati A, Ongaro A, Fini M, et al. Adenosine analogs and electromagnetic fields inhibit prostaglandin E2 release in bovine synovial fibroblasts. Osteoarthr Cartil. 2009;17(2):252–262. doi: 10.1016/j.joca.2008.06.002.
    1. Ongaro A, Pellati A, Setti S, Masieri FF, Aquila G, Fini M, et al. Electromagnetic fields counteract IL-1beta activity during chondrogenesis of bovine mesenchymal stem cells. J Tissue Eng Regen Med. 2015;9(12):E229–E238. doi: 10.1002/term.1671.
    1. Ongaro A, Varani K, Masieri FF, Pellati A, Massari L, Cadossi R, et al. Electromagnetic fields (EMFs) and adenosine receptors modulate prostaglandin E (2) and cytokine release in human osteoarthritic synovial fibroblasts. J Cell Physiol. 2012;227(6):2461–2469. doi: 10.1002/jcp.22981.
    1. Raghothaman D, Leong MF, Lim TC, Toh JK, Wan AC, Yang Z, et al. Engineering cell matrix interactions in assembled polyelectrolyte fiber hydrogels for mesenchymal stem cell chondrogenesis. Biomaterials. 2014;35(9):2607–2616. doi: 10.1016/j.biomaterials.2013.12.008.
    1. Yang Z, Wu Y, Li C, Zhang T, Zou Y, Hui JH, et al. Improved mesenchymal stem cells attachment and in vitro cartilage tissue formation on chitosan-modified poly(L-lactide-co-epsilon-caprolactone) scaffold. Tissue Eng Part A. 2012;18(3–4):242–251. doi: 10.1089/ten.tea.2011.0315.
    1. Yin L, Wu Y, Yang Z, Denslin V, Ren X, Tee CA, et al. Characterization and application of size-sorted zonal chondrocytes for articular cartilage regeneration. Biomaterials. 2018;165:66–78. doi: 10.1016/j.biomaterials.2018.02.050.
    1. Qazi TH, Mooney DJ, Duda GN, Geissler S. Biomaterials that promote cell-cell interactions enhance the paracrine function of MSCs. Biomaterials. 2017;140:103–114. doi: 10.1016/j.biomaterials.2017.06.019.
    1. Goldring MB, Tsuchimochi K, Ijiri K. The control of chondrogenesis. J Cell Biochem. 2006;97(1):33–44. doi: 10.1002/jcb.20652.
    1. Li Z, Kupcsik L, Yao SJ, Alini M, Stoddart MJ. Mechanical load modulates chondrogenesis of human mesenchymal stem cells through the TGF-beta pathway. J Cell Mol Med. 2010;14(6A):1338–1346. doi: 10.1111/j.1582-4934.2009.00780.x.
    1. Zhu Y, Wang Y, Zhao B, Niu X, Hu B, Li Q, 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.
    1. Vonk LA, van Dooremalen SFJ, Liv N, Klumperman J, Coffer PJ, Saris DBF, et al. Mesenchymal stromal/stem cell-derived extracellular vesicles promote human cartilage regeneration in vitro. Theranostics. 2018;8(4):906–920. doi: 10.7150/thno.20746.
    1. Glyn-Jones S, Palmer AJ, Agricola R, Price AJ, Vincent TL, Weinans H, et al. Osteoarthritis. Lancet. 2015;386(9991):376–387. doi: 10.1016/S0140-6736(14)60802-3.
    1. Hunziker EB, Rosenberg LC. Repair of partial-thickness defects in articular cartilage: cell recruitment from the synovial membrane. J Bone Joint Surg Am. 1996;78(5):721–733. doi: 10.2106/00004623-199605000-00012.
    1. Zhang W, Chen J, Tao J, Jiang Y, Hu C, Huang L, et al. The use of type 1 collagen scaffold containing stromal cell-derived factor-1 to create a matrix environment conducive to partial-thickness cartilage defects repair. Biomaterials. 2013;34(3):713–723. doi: 10.1016/j.biomaterials.2012.10.027.
    1. Schmitt B, Ringe J, Haupl T, Notter M, Manz R, Burmester GR, et al. BMP2 initiates chondrogenic lineage development of adult human mesenchymal stem cells in high-density culture. Differentiation. 2003;71(9–10):567–577. doi: 10.1111/j.1432-0436.2003.07109003.x.
    1. Jiang Y, Chen LK, Zhu DC, Zhang GR, Guo C, Qi YY, et al. The inductive effect of bone morphogenetic protein-4 on chondral-lineage differentiation and in situ cartilage repair. Tissue Eng Part A. 2010;16(5):1621–1632. doi: 10.1089/ten.tea.2009.0681.
    1. Taylor DK, Meganck JA, Terkhorn S, Rajani R, Naik A, O'Keefe RJ, et al. Thrombospondin-2 influences the proportion of cartilage and bone during fracture healing. J Bone Miner Res. 2009;24(6):1043–1054. doi: 10.1359/jbmr.090101.
    1. Jeong SY, Kim DH, Ha J, Jin HJ, Kwon SJ, Chang JW, et al. Thrombospondin-2 secreted by human umbilical cord blood-derived mesenchymal stem cells promotes chondrogenic differentiation. Stem Cells. 2013;31(10):2136–2148. doi: 10.1002/stem.1471.
    1. Jeong SY, Ha J, Lee M, Jin HJ, Kim DH, Choi SJ, et al. Autocrine action of thrombospondin-2 determines the chondrogenic differentiation potential and suppresses hypertrophic maturation of human umbilical cord blood-derived mesenchymal stem cells. Stem Cells. 2015;33(11):3291–3303. doi: 10.1002/stem.2120.
    1. Fiedler J, Roderer G, Gunther KP, Brenner RE. BMP-2, BMP-4, and PDGF-bb stimulate chemotactic migration of primary human mesenchymal progenitor cells. J Cell Biochem. 2002;87(3):305–312. doi: 10.1002/jcb.10309.
    1. Lamacchia C, Rodriguez E, Palmer G, Vigne S, Martin P, Talabot-Ayer D, et al. Articular inflammation is controlled by myeloid cell-derived interleukin 1 receptor antagonist during the acute phase of arthritis in mice. Ann Rheum Dis. 2012;71(2):281–287. doi: 10.1136/annrheumdis-2011-200429.
    1. Elsaid KA, Zhang L, Shaman Z, Patel C, Schmidt TA, Jay GD. The impact of early intra-articular administration of interleukin-1 receptor antagonist on lubricin metabolism and cartilage degeneration in an anterior cruciate ligament transection model. Osteoarthr Cartil. 2015;23(1):114–121. doi: 10.1016/j.joca.2014.09.006.
    1. 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.
    1. Wu J, Kuang L, Chen C, Yang J, Zeng WN, Li T, 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. doi: 10.1016/j.biomaterials.2019.03.022.
    1. Mao G, Hu S, Zhang Z, Wu P, Zhao X, Lin R, et al. Exosomal miR-95-5p regulates chondrogenesis and cartilage degradation via histone deacetylase 2/8. J Cell Mol Med. 2018;22(11):5354–5366. doi: 10.1111/jcmm.13808.
    1. Zhang Y, Yan J, Xu H, Yang Y, Li W, Wu H, et al. Extremely low frequency electromagnetic fields promote mesenchymal stem cell migration by increasing intracellular Ca (2+) and activating the FAK/Rho GTPases signaling pathways in vitro. Stem Cell Res Ther. 2018;9(1):143. doi: 10.1186/s13287-018-0883-4.
    1. Crocetti S, Beyer C, Schade G, Egli M, Frohlich J, Franco-Obregon A. Low intensity and frequency pulsed electromagnetic fields selectively impair breast cancer cell viability. PLoS One. 2013;8(9):e72944. doi: 10.1371/journal.pone.0072944.

Source: PubMed

3
Tilaa