Enhancement of mesenchymal stem cell chondrogenesis with short-term low intensity pulsed electromagnetic fields

Dinesh Parate, Alfredo Franco-Obregón, Jürg Fröhlich, Christian Beyer, Azlina A Abbas, Tunku Kamarul, James H P Hui, Zheng Yang, Dinesh Parate, Alfredo Franco-Obregón, Jürg Fröhlich, Christian Beyer, Azlina A Abbas, Tunku Kamarul, James H P Hui, Zheng Yang

Abstract

Pulse electromagnetic fields (PEMFs) have been shown to recruit calcium-signaling cascades common to chondrogenesis. Here we document the effects of specified PEMF parameters over mesenchymal stem cells (MSC) chondrogenic differentiation. MSCs undergoing chondrogenesis are preferentially responsive to an electromagnetic efficacy window defined by field amplitude, duration and frequency of exposure. Contrary to conventional practice of administering prolonged and repetitive exposures to PEMFs, optimal chondrogenic outcome is achieved in response to brief (10 minutes), low intensity (2 mT) exposure to 6 ms bursts of magnetic pulses, at 15 Hz, administered only once at the onset of chondrogenic induction. By contrast, repeated exposures diminished chondrogenic outcome and could be attributed to calcium entry after the initial induction. Transient receptor potential (TRP) channels appear to mediate these aspects of PEMF stimulation, serving as a conduit for extracellular calcium. Preventing calcium entry during the repeated PEMF exposure with the co-administration of EGTA or TRP channel antagonists precluded the inhibition of differentiation. This study highlights the intricacies of calcium homeostasis during early chondrogenesis and the constraints that are placed on PEMF-based therapeutic strategies aimed at promoting MSC chondrogenesis. The demonstrated efficacy of our optimized PEMF regimens has clear clinical implications for future regenerative strategies for cartilage.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Effects of PEMF amplitude (A) and exposure duration (B) on MSC chondrogenesis. Real-time PCR analysis of cartilaginous markers expression after 7 days of differentiation was normalized to GAPDH and presented as fold-changes relative to levels in undifferentiated MSC. (C) Quantification of cartilaginous extracellular matrix macromolecules (Col 2 and sGAG) generated after 21 days of chondrogenic differentiation of MSC subjected to distinct PEMF parameters. All data shown are mean ± SD, n = 6 from 2 independent experiments. *Denotes significant increase, or decrease, compared to non-PEMF control. #Denotes significant decrease compared to 2 mT (A), or 10 min (B) PEMF exposure.
Figure 2
Figure 2
Dosage effects of PEMFs over MSC chondrogenesis. (A) MSCs were exposed once (1x), twice (2x) or thrice (3x) per week. (B) MSCs were subjected to either a single exposure on day 1 of chondrogenic induction (1x) or once per week for 3 weeks (3x). Real-time PCR analysis of cartilaginous markers expression at 7 (A) or 21 days (B) after the induction of differentiation was normalized to GAPDH and presented as fold-changes relative to level in undifferentiated MSCs. (C) Quantification of cartilaginous ECM macromolecules generated during chondrogenic differentiation of MSCs in response to distinct PEMF dosing as indicated. MSC pellets were subjected to either a single PEMF exposure given on day 1 of chondrogenic induction (1x) once per week for 3 weeks (3x), or thrice weekly for 3 weeks (9x). Data represents the mean ± SD, n = 6 from 2 independent experiments. *Denotes significant increase compare to non-PEMF (0 mT) control. #Denotes significant decrease compared to single PEMF (1x) exposure.
Figure 3
Figure 3
Histological analysis of pellets exposed to PEMFs of distinct amplitude, duration and dosage. Pellets were harvested at day 21, sectioned and subjected to Safranin O or type II collagen immunohistochemistry staining. Images presented were represenation of n = 3, taken at 100× magnification.
Figure 4
Figure 4
Investigation of calcium entry pathways implicated in the PEMF-effect. (A) Involvement of Ca2+ influx in mediating the effects of PEMF-induced MSC chondrogenic differentiation. MSCs were exposed for 10 min at 2 mT alone (control, white bars), or in the presence of 2 mM EGTA (dark grey bars) or 5 mM CaCl2 (hatched bars) transiently added to the culture media. EGTA and CaCl2 were included to the bathing media 10 min before exposure and replaced with age-matched media control cultures 10 min after exposure. (B) Involvement of candidate calcium channels in mediating the effect of PEMs over MSC chondrogenic differentiation. Control MSC chondrogenic differentiation medium (white bars) was supplemented with Nifedipine (1 µM, light grey bars), Ruthenium Red (RR, 10 µM, black bars), or 2-APB (100 µM, dark grey bars) 10 min before exposure and replaced with age-matched media control cultures 10 min after exposure. Real-time PCR analysis was performed on day 7 of differentiation. Data represent the means ± SD, n = 6 from 2 independent experiments. *Denotes significant increase, or decrease, compared to non-PEMF (0 mT) control. #Denotes significant decrease relativeto 2 mT PEMF treatment.
Figure 5
Figure 5
Expression profiles of TRPC1 and TRPV4 in response to determined PEMF efficacy window regulating MSC chondrogenesis. Real-time PCR analysis of TRPC1 and V4 exposed to different (A) intensities, (B) durations, and (C) dosages of PEMFs. Data represent the means ± SD, n = 6 from 2 independent experiments. *Denotes significant increase, or decrease, compared to non-PEMF (0 mT) control. #Denotes significant decrease relative to 2 mT (A), 10 min (B), or single (1x, C) PEMF treatment.
Figure 6
Figure 6
(A) MSC chondrogenic differentiation in response to multiple exposures to PEMFs or exogenously elevated calcium. MSCs were subjected to either PEMF stimulation alone (white bars) or with transient supplementation of CaCl2 alone (5 mM; hatched bars), once (1x), twice (2x) or thrice (3x) in a week. Dotted lines refers to expression level of non-treated controls. Real-time PCR analysis was performed on day 7 of chondrogenic differentiation. *Denotes significant increase relative to non-PEMF (0 mT) control. # and + denote significant differences relative to respective single (1x) exposure (white and hatched bars, respectively). P = PEMF treatment, Ca = CaCl2 supplementation. (B) MSC chondrogenic differentiation in response to multiple exposures to PEMFs alone (white bars) or in combination with calcium chelator (EGTA) or TRP channel antagonists. EGTA (2 mM; dark grey bars; “E”), Ruthenium Red (10 µM; RR, black bars; “R”) or 2-APB (100 µM; light grey bars; “C”) was added to the MSC differentiation medium during PEMF expoure applied once (1x), twice (2) or thrice (3x) per week. EGTA, RR and 2-APB were included 10 min before exposure and replaced with media harvested from age-matched chondrogenic control cultures 10 min after exposure. *Denotes significant increase compare to non-PEMF (0 mT) control. #Denotes significant decrease compared to single PEMF exposure (1x). +Denotes significant difference compared to respective PEMF control (white bar). P = PEMF treatment, E = EGTA, R = Ruthenium Red (RR), C = 2-APB. Data shown are means ± SD, n = 6 from 2 independent experiments.
Figure 7
Figure 7
Quantification of cartilaginous ECM macromolecules generated by chondrogenically differentiated MSC in response to single or three weekly PEMF exposures alone (white bars) or in combination with calcium chelator (EGTA) or TRP channel antagonists as indicated. EGTA (2 mM; dark grey bars; “E”), Ruthenium Red (10 µM; RR, black bars; “R”) and 2-APB (100 µM; light grey bars; “C”) were included once during single PEMF exposures, or twice during the second and third PEMF exposure. EGTA, RR and 2-APB were added 10 min before exposure and replaced with media harvested from age-matched chondrogenic control cultures 10 min after exposure. *Denotes significant increase compare to non-PEMF (0 mT) control. #Denotes significant decrease compared to single PEMF exposure (1x). +Denotes significant difference compared to respective PEMF control (white bar). P = PEMF treatment, E = EGTA, R = Ruthenium Red (RR), C = 2-APB. Data represent means ± SD, n = 3.

References

    1. Poole, A. R. et al. Composition and structure of articular cartilage: a template for tissue repair. Clin Orthop Relat Res S26–33 (2001).
    1. Goldring MB, Goldring SR. Osteoarthritis. J Cell Physiol. 2007;213:626–34. doi: 10.1002/jcp.21258.
    1. Ge Z, et al. Osteoarthritis and therapy. Arthritis Rheum. 2006;55:493–500. doi: 10.1002/art.21994.
    1. Buckwalter JA, Mankin HJ. Articular cartilage: degeneration and osteoarthritis, repair, regeneration, and transplantation. Instr Course Lect. 1998;47:487–504.
    1. Vasara, A.I. et al. Indentation stiffness of repair tissue after autologous chondrocyte transplantation. Clin Orthop Relat Res, 233–42 (2005).
    1. Wakitani S, et al. Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage. J Bone Joint Surg Am. 1994;76:579–92. doi: 10.2106/00004623-199404000-00013.
    1. Wakitani S, et al. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002;10:199–206. doi: 10.1053/joca.2001.0504.
    1. Kuroda R, et al. Treatment of a full-thickness articular cartilage defect in the femoral condyle of an athlete with autologous bone-marrow stromal cells. Osteoarthritis Cartilage. 2007;15:226–31. doi: 10.1016/j.joca.2006.08.008.
    1. Pittenger MF, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143.
    1. Goldring MB, Tsuchimochi K, Ijiri K. The control of chondrogenesis. J Cell Biochem. 2006;97:33–44. doi: 10.1002/jcb.20652.
    1. Mahmoudifar N, Doran PM. Chondrogenesis and cartilage tissue engineering: the longer road to technology development. Trends Biotechnol. 2012;30:166–76. doi: 10.1016/j.tibtech.2011.09.002.
    1. Woods A, Wang G, Beier F. Regulation of chondrocyte differentiation by the actin cytoskeleton and adhesive interactions. J Cell Physiol. 2007;213:1–8. doi: 10.1002/jcp.21110.
    1. DeLise AM, Fischer L, Tuan RS. Cellular interactions and signaling in cartilage development. Osteoarthritis Cartilage. 2000;8:309–34. doi: 10.1053/joca.1999.0306.
    1. Toh WS, Spector M, Lee EH, Cao T. Biomaterial-mediated delivery of microenvironmental cues for repair and regeneration of articular cartilage. Mol Pharm. 2011;8:994–1001. doi: 10.1021/mp100437a.
    1. Klein TJ, Malda J, Sah RL, Hutmacher DW. Tissue engineering of articular cartilage with biomimetic zones. Tissue Eng Part B Rev. 2009;15:143–57. doi: 10.1089/ten.teb.2008.0563.
    1. Raghothaman D, et al. Engineering cell matrix interactions in assembled polyelectrolyte fiber hydrogels for mesenchymal stem cell chondrogenesis. Biomaterials. 2014;35:2607–16. doi: 10.1016/j.biomaterials.2013.12.008.
    1. Responte DJ, Lee JK, Hu JC, Athanasiou KA. Biomechanics-driven chondrogenesis: from embryo to adult. FASEB J. 2012;26:3614–24. doi: 10.1096/fj.12-207241.
    1. Mouw JK, Connelly JT, Wilson CG, Michael KE, Levenston ME. Dynamic compression regulates the expression and synthesis of chondrocyte-specific matrix molecules in bone marrow stromal cells. Stem Cells. 2007;25:655–63. doi: 10.1634/stemcells.2006-0435.
    1. Grad S, Eglin D, Alini M, Stoddart MJ. Physical stimulation of chondrogenic cells in vitro: a review. Clin Orthop Relat Res. 2011;469:2764–72. doi: 10.1007/s11999-011-1819-9.
    1. Cheing GL, Li X, Huang L, Kwan RL, Cheung KK. Pulsed electromagnetic fields (PEMF) promote early wound healing and myofibroblast proliferation in diabetic rats. Bioelectromagnetics. 2014;35:161–9. doi: 10.1002/bem.21832.
    1. Steward AJ, Kelly DJ, Wagner DR. The role of calcium signalling in the chondrogenic response of mesenchymal stem cells to hydrostatic pressure. Eur Cell Mater. 2014;28:358–71. doi: 10.22203/eCM.v028a25.
    1. Pingguan-Murphy B, El-Azzeh M, Bader DL, Knight MM. Cyclic compression of chondrocytes modulates a purinergic calcium signalling pathway in a strain rate- and frequency-dependent manner. J Cell Physiol. 2006;209:389–97. doi: 10.1002/jcp.20747.
    1. Eijkelkamp N, Quick K, Wood JN. Transient receptor potential channels and mechanosensation. Annu Rev Neurosci. 2013;36:519–46. doi: 10.1146/annurev-neuro-062012-170412.
    1. Matta C, Zakany R. Calcium signalling in chondrogenesis: implications for cartilage repair. Front Biosci (Schol Ed) 2013;5:305–24. doi: 10.2741/S374.
    1. Pall ML. Electromagnetic fields act via activation of voltage-gated calcium channels to produce beneficial or adverse effects. J Cell Mol Med. 2013;17:958–65. doi: 10.1111/jcmm.12088.
    1. Ross CL, et al. The effect of low-frequency electromagnetic field on human bone marrow stem/progenitor cell differentiation. Stem Cell Res. 2015;15:96–108. doi: 10.1016/j.scr.2015.04.009.
    1. Petecchia L, et al. Electro-magnetic field promotes osteogenic differentiation of BM-hMSCs through a selective action on Ca(2+)-related mechanisms. Sci Rep. 2015;5 doi: 10.1038/srep13856.
    1. Schmidt-Rohlfing B, Silny J, Woodruff S, Gavenis K. Effects of pulsed and sinusoid electromagnetic fields on human chondrocytes cultivated in a collagen matrix. Rheumatol Int. 2008;28:971–7. doi: 10.1007/s00296-008-0565-0.
    1. Vincenzi F, et al. Pulsed electromagnetic fields increased the anti-inflammatory effect of A(2)A and A(3) adenosine receptors in human T/C-28a2 chondrocytes and hFOB 1.19 osteoblasts. PLoS One. 2013;8 doi: 10.1371/journal.pone.0065561.
    1. De Mattei M, et al. Effects of pulsed electromagnetic fields on human articular chondrocyte proliferation. Connect Tissue Res. 2001;42:269–79. doi: 10.3109/03008200109016841.
    1. Sakai A, Suzuki K, Nakamura T, Norimura T, Tsuchiya T. Effects of pulsing electromagnetic fields on cultured cartilage cells. Int Orthop. 1991;15:341–6. doi: 10.1007/BF00186874.
    1. Hilz FM, et al. Influence of extremely low frequency, low energy electromagnetic fields and combined mechanical stimulation on chondrocytes in 3-D constructs for cartilage tissue engineering. Bioelectromagnetics. 2014;35:116–28. doi: 10.1002/bem.21822.
    1. Boopalan PR, Arumugam S, Livingston A, Mohanty M, Chittaranjan S. Pulsed electromagnetic field therapy results in healing of full thickness articular cartilage defect. Int Orthop. 2011;35:143–8. doi: 10.1007/s00264-010-0994-8.
    1. Mueller MB, Tuan RS. Functional characterization of hypertrophy in chondrogenesis of human mesenchymal stem cells. Arthritis Rheum. 2008;58:1377–88. doi: 10.1002/art.23370.
    1. Chen CH, et al. Electromagnetic fields enhance chondrogenesis of human adipose-derived stem cells in a chondrogenic microenvironment in vitro. J Appl Physiol (1985) 2013;114:647–55. doi: 10.1152/japplphysiol.01216.2012.
    1. Lim, H. L. et al. Dynamic Electromechanical Hydrogel Matrices for Stem Cell Culture. Adv Funct Mater21 (2011).
    1. Wang J, et al. Pulsed electromagnetic field may accelerate in vitro endochondral ossification. Bioelectromagnetics. 2015;36:35–44. doi: 10.1002/bem.21882.
    1. Mayer-Wagner S, et al. Effects of low frequency electromagnetic fields on the chondrogenic differentiation of human mesenchymal stem cells. Bioelectromagnetics. 2011;32:283–90. doi: 10.1002/bem.20633.
    1. Esposito M, et al. Differentiation of human umbilical cord-derived mesenchymal stem cells, WJ-MSCs, into chondrogenic cells in the presence of pulsed electromagnetic fields. In Vivo. 2013;27:495–500.
    1. Ongaro A, et al. Electromagnetic fields counteract IL-1beta activity during chondrogenesis of bovine mesenchymal stem cells. J Tissue Eng Regen Med. 2015;9:E229–38. doi: 10.1002/term.1671.
    1. Spaas JH, et al. Chondrogenic Priming at Reduced Cell Density Enhances Cartilage Adhesion of Equine Allogeneic MSCs - a Loading Sensitive Phenomenon in an Organ Culture Study with 180 Explants. Cell Physiol Biochem. 2015;37:651–65. doi: 10.1159/000430384.
    1. Berg H. Problems of weak electromagnetic field effects in cell biology. Bioelectrochem Bioenerg. 1999;48:355–60. doi: 10.1016/S0302-4598(99)00012-4.
    1. Crocetti S, et al. Low intensity and frequency pulsed electromagnetic fields selectively impair breast cancer cell viability. PLoS One. 2013;8 doi: 10.1371/journal.pone.0072944.
    1. O’Conor CJ, Case N, Guilak F. Mechanical regulation of chondrogenesis. Stem Cell Res Ther. 2013;4 doi: 10.1186/scrt211.
    1. Sun LY, et al. Effect of pulsed electromagnetic field on the proliferation and differentiation potential of human bone marrow mesenchymal stem cells. Bioelectromagnetics. 2009;30:251–60. doi: 10.1002/bem.20472.
    1. Fodor J, et al. Store-operated calcium entry and calcium influx via voltage-operated calcium channels regulate intracellular calcium oscillations in chondrogenic cells. Cell Calcium. 2013;54:1–16. doi: 10.1016/j.ceca.2013.03.003.
    1. San Antonio JD, Tuan RS. Chondrogenesis of limb bud mesenchyme in vitro: stimulation by cations. Dev Biol. 1986;115:313–24. doi: 10.1016/0012-1606(86)90252-6.
    1. Matta C, et al. Cytosolic free Ca2+ concentration exhibits a characteristic temporal pattern during in vitro cartilage differentiation: a possible regulatory role of calcineurin in Ca-signalling of chondrogenic cells. Cell Calcium. 2008;44:310–23. doi: 10.1016/j.ceca.2007.12.010.
    1. Argentaro A, et al. A SOX9 defect of calmodulin-dependent nuclear import in campomelic dysplasia/autosomal sex reversal. J Biol Chem. 2003;278:33839–47. doi: 10.1074/jbc.M302078200.
    1. McCullen SD, et al. Application of low-frequency alternating current electric fields via interdigitated electrodes: effects on cellular viability, cytoplasmic calcium, and osteogenic differentiation of human adipose-derived stem cells. Tissue Eng Part C Methods. 2010;16:1377–86. doi: 10.1089/ten.tec.2009.0751.
    1. Xu J, Wang W, Clark CC, Brighton CT. Signal transduction in electrically stimulated articular chondrocytes involves translocation of extracellular calcium through voltage-gated channels. Osteoarthritis Cartilage. 2009;17:397–405. doi: 10.1016/j.joca.2008.07.001.
    1. Kawano S, et al. Characterization of Ca(2+) signaling pathways in human mesenchymal stem cells. Cell Calcium. 2002;32:165–74. doi: 10.1016/S0143416002001240.
    1. Curtis TM, Scholfield CN. Nifedipine blocks Ca2+ store refilling through a pathway not involving L-type Ca2+ channels in rabbit arteriolar smooth muscle. J Physiol. 2001;532:609–23. doi: 10.1111/j.1469-7793.2001.0609e.x.
    1. Wen L, et al. L-type calcium channels play a crucial role in the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells. Biochem Biophys Res Commun. 2012;424:439–45. doi: 10.1016/j.bbrc.2012.06.128.
    1. Zhang J, Li M, Kang ET, Neoh KG. Electrical stimulation of adipose-derived mesenchymal stem cells in conductive scaffolds and the roles of voltage-gated ion channels. Acta Biomater. 2016;32:46–56. doi: 10.1016/j.actbio.2015.12.024.
    1. Lin SS, et al. Cav3.2 T-type calcium channel is required for the NFAT-dependent Sox9 expression in tracheal cartilage. Proc Natl Acad Sci USA. 2014;111:E1990–8. doi: 10.1073/pnas.1323112111.
    1. Guilak F, Leddy HA, Liedtke W. Transient receptor potential vanilloid 4: The sixth sense of the musculoskeletal system? Ann N Y Acad Sci. 2010;1192:404–9. doi: 10.1111/j.1749-6632.2010.05389.x.
    1. Maroto R, et al. TRPC1 forms the stretch-activated cation channel in vertebrate cells. Nat Cell Biol. 2005;7:179–85. doi: 10.1038/ncb1218.
    1. Eleswarapu SV, Athanasiou KA. TRPV4 channel activation improves the tensile properties of self-assembled articular cartilage constructs. Acta Biomater. 2013;9:5554–61. doi: 10.1016/j.actbio.2012.10.031.
    1. Phan MN, et al. Functional characterization of TRPV4 as an osmotically sensitive ion channel in porcine articular chondrocytes. Arthritis Rheum. 2009;60:3028–37. doi: 10.1002/art.24799.
    1. Hung CT. Transient receptor potential vanilloid 4 channel as an important modulator of chondrocyte mechanotransduction of osmotic loading. Arthritis Rheum. 2010;62:2850–1. doi: 10.1002/art.27617.
    1. Kurth F, et al. Transient receptor potential vanilloid 2-mediated shear-stress responses in C2C12 myoblasts are regulated by serum and extracellular matrix. FASEB J. 2015;29:4726–37. doi: 10.1096/fj.15-275396.
    1. Clapham DE, Runnels LW, Strubing C. The TRP ion channel family. Nat Rev Neurosci. 2001;2:387–96. doi: 10.1038/35077544.
    1. Somogyi CS, et al. Polymodal Transient Receptor Potential Vanilloid (TRPV) Ion Channels in Chondrogenic Cells. Int J Mol Sci. 2015;16:18412–38. doi: 10.3390/ijms160818412.
    1. Muramatsu S, et al. Functional gene screening system identified TRPV4 as a regulator of chondrogenic differentiation. J Biol Chem. 2007;282:32158–67. doi: 10.1074/jbc.M706158200.
    1. Gavenis K, et al. Expression of ion channels of the TRP family in articular chondrocytes from osteoarthritic patients: changes between native and in vitro propagated chondrocytes. Mol Cell Biochem. 2009;321:135–43. doi: 10.1007/s11010-008-9927-x.
    1. Torossian F, Bisson A, Vannier JP, Boyer O, Lamacz M. TRPC expression in mesenchymal stem cells. Cell Mol Biol Lett. 2010;15:600–10. doi: 10.2478/s11658-010-0031-3.
    1. Lievremont JP, Bird GS, Putney JW., Jr. Mechanism of inhibition of TRPC cation channels by 2-aminoethoxydiphenylborane. Mol Pharmacol. 2005;68:758–62.
    1. Yang Z, 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;18:242–51. doi: 10.1089/ten.tea.2011.0315.
    1. Winegar BD, Haws CM, Lansman JB. Subconductance block of single mechanosensitive ion channels in skeletal muscle fibers by aminoglycoside antibiotics. J Gen Physiol. 1996;107:433–43. doi: 10.1085/jgp.107.3.433.

Source: PubMed

3
Subscribe