Intra-articular delivery of adipose derived stromal cells attenuates osteoarthritis progression in an experimental rabbit model

Giovanna Desando, Carola Cavallo, Federica Sartoni, Lucia Martini, Annapaola Parrilli, Francesca Veronesi, Milena Fini, Roberto Giardino, Andrea Facchini, Brunella Grigolo, Giovanna Desando, Carola Cavallo, Federica Sartoni, Lucia Martini, Annapaola Parrilli, Francesca Veronesi, Milena Fini, Roberto Giardino, Andrea Facchini, Brunella Grigolo

Abstract

Introduction: Cell therapy is a rapidly growing area of research for the treatment of osteoarthritis (OA). This work is aimed to investigate the efficacy of intra-articular adipose-derived stromal cell (ASC) injection in the healing process on cartilage, synovial membrane and menisci in an experimental rabbit model.

Methods: The induction of OA was performed surgically through bilateral anterior cruciate ligament transection (ACLT) to achieve eight weeks from ACLT a mild grade of OA. A total of 2×10⁶ and 6×10⁶ autologous ASCs isolated from inguinal fat, expanded in vitro and suspended in 4% rabbit serum albumin (RSA) were delivered in the hind limbs; 4% RSA was used as the control. Local bio-distribution of the cells was verified by injecting chloro-methyl-benzamido-1,1'-dioctadecyl-3,3,3'3'-tetra-methyl-indo-carbocyanine per-chlorate (CM-Dil) labeled ASCs in the hind limbs. Cartilage and synovial histological sections were scored by Laverty's scoring system to assess the severity of the pathology. Protein expression of some extracellular matrix molecules (collagen I and II), catabolic (metalloproteinase-1 and -3) and inflammatory (tumor necrosis factor- α) markers were detected by immunohistochemistry. Assessments were carried out at 16 and 24 weeks.

Results: Labeled-ASCs were detected unexpectedly in the synovial membrane and medial meniscus but not in cartilage tissue at 3 and 20 days from ASC-treatment. Intra-articular ASC administration decreases OA progression and exerts a healing contribution in the treated animals in comparison to OA and 4% RSA groups.

Conclusions: Our data reveal a healing capacity of ASCs in promoting cartilage and menisci repair and attenuating inflammatory events in synovial membrane inhibiting OA progression. On the basis of the local bio-distribution findings, the benefits obtained by ASC treatment could be due to a trophic mechanism of action by the release of growth factors and cytokines.

Figures

Figure 1
Figure 1
Degenerative changes in different articular compartments in sham and OA groups at eight weeks. (A, B) India ink staining of medial femoral condyle (MFC) of representative specimens. Arrows, fibrillation processes (C, D) Safranin-O/Fast Green staining of cartilage tissue of representative specimens. Arrows: Fibrillation and delamination processes. (E, F) Hematoxylin/Eosin staining of synovial membrane of representative specimens. Black arrows, Thickening of lining layer; Red arrows, Inflammatory cells. (G, H) Safranin-O/Fast Green staining of medial meniscus of representative specimens. Scale bars in C, D = 1 mm; E-H = 100 μm.
Figure 2
Figure 2
Chondrogenic and osteogenic differentiation of rabbit adipose stem cells (ASCs) in vitro. (A) Chondrogenic and osteogenic differentiation of rabbit ASCs labeled with CM-Dil in vitro. (B) Growth curves of unlabeled and labeled ASCs (C).
Figure 3
Figure 3
Biodistribution of CM-Dil labeled ASCs. Detection of labeled-ASCs in synovial lining layer (A-D) and medial meniscus (E-H) of representative specimens at 3 and 20 days from ASC administration under bright field (A, C, E, G) and epi-fluorescence (B, D, F, H). Blue staining = nuclei; Red staining = CM-Dil labeled-ASCs. Scale bars = 500 μm.
Figure 4
Figure 4
Quantitative assessment of disease progression in the different groups. (A) Fibrillation index. (B) Cartilage thickness. Data are reported in terms of mean ± SD. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 5
Figure 5
Histological evaluation of medial femoral condyle in OA, 4% RSA and ASC-treated groups. (A) Safranin-O/Fast Green staining of representative specimens. Black arrows, fibrillation and delamination processes. Red arrows, Erosion processes. Scale bars = 1,000 μm. (B) Laverty's score. Data are reported in terms of 95% confidence intervals. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 6
Figure 6
Histological evaluation of synovial membrane in OA, 4% RSA and ASC-treated groups. (A) H/E staining of representative specimens. Scale bars = 100 μm. (B) Laverty's score. Data are reported in terms of the 95% confidence intervals. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 7
Figure 7
Histological evaluation of menisci in OA, 4% RSA and ASC-treated groups. (A) Safranin-O/Fast Green staining of representative specimens. Scale bars = 100 μm. (B) Numbers of cluster formation. All the data are reported as 95% confidence intervals. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 8
Figure 8
Immunohistochemical analysis for collagen II and I in cartilage. Photomicrographs of representative specimens evaluated for type II (A) and I (B) collagens in medial femoral condyle of OA, 4% RSA and ASC-treated groups. Data are reported as mean ± SD. Scale bars = 100 μm. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 9
Figure 9
Immunohistochemical analysis for MMP-1 and TNF-α in cartilage. Photomicrographs of representative specimens evaluated for MMP-1 (A) and TNF-α (B) in medial femoral condyle of OA, 4% RSA and ASC-treated groups. Data are reported as mean ± SD. Scale bars = 100 μm. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 10
Figure 10
Immunohistochemical analysis for MMP-1 and TNF-α in synovial membrane. Photomicrographs of representative specimens evaluated for MMP-1 (A) and TNF-α (B) in synovial membrane of OA, 4% RSA and ASC-treated groups. Data are reported as mean ± SD. Scale bars = 100 μm. Statistical values of at least P < 0.01 were observed in all the comparisons.
Figure 11
Figure 11
Immunohistochemical analysis for MMP-1 and TNF-α in menisci. Photomicrographs of representative specimens evaluated for MMP-1 (A) and TNF-α (B) in medial menisci of OA, 4% RSA and ASC-treated groups. Data are reported as mean ± SD. Scale bars = 100 μm. Statistical values of at least P < 0.01 were observed in all the comparisons.

References

    1. Scott JC, Lethbridge-Cejku M, Hochberg MC. In: Osteoarthritis: Clinical and Experimental Aspects. 1. Reginster JY, Pelletier JP, Marte-Pelletier J, Henrotin Y, editor. Vol. 1. Munich, Germany: Springer; 1999. Epidemiology and economic consequences of osteoarthritis; pp. 20–52.
    1. Goldring MB, Goldring SR. Osteoarthritis. J Cell Physiol. 2007;213:626–634. doi: 10.1002/jcp.21258.
    1. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum. 2012;64:1697–1707. doi: 10.1002/art.34453.
    1. Okada Y, Shinmei M, Tanaka O, Naka K, Kimura A, Nakanishi I, Bayliss MT, Iwata K, Nagase H. Localization of matrix metalloproteinase 3 (stromelysin) in osteoarthritic cartilage and synovium. Lab Invest. 1992;66:680–690.
    1. Goldring SR, Goldring MB. The role of cytokines in cartilage matrix degeneration in osteoarthritis. Clin Orthop Relat Res. 2004;427(suppl):S27–S36.
    1. Goldring MB, Marcu KB. Cartilage homeostasis in health and rheumatic diseases. Arthritis Res Ther. 2009;11:224. doi: 10.1186/ar2592.
    1. Hunziker EB. Articular cartilage repair: basic science and clinical progress. A review of the current status and prospects. Osteoarthritis Cartilage. 2002;10:432–463. doi: 10.1053/joca.2002.0801.
    1. Recommendations for the medical management of osteoarthritis of the hip and knee: 2000 update. American College of Rheumatology Subcommittee on Osteoarthritis Guidelines. Arthritis Rheum. 2000;43:1905–1915.
    1. Zhang W, Moskowitz RW, Nuki G, Abramson S, Altman RD, Arden N, Bierma-Zeinstra S, Brandt KD, Croft P, Doherty M, Dougados M, Hochberg M, Hunter DJ, Kwoh K, Lohmander LS, Tugwell P. OARSI recommendations for the management of hip and knee osteoarthritis, part I: critical appraisal of existing treatment guidelines and systematic review of current research evidence. Osteoarthritis Cartilage. 2007;15:981–1000. doi: 10.1016/j.joca.2007.06.014.
    1. Gibofsky A, Barkin RL. Chronic pain of osteoarthritis: considerations for selecting an extended-release opioid analgesic. Am J Ther. 2008;15:241–255. doi: 10.1097/MJT.0b013e3181727f68.
    1. Caplan AI, Bruder SP. Mesenchymal stem cells: building blocks for molecular medicine in the 21st century. Trends Mol Med. 2001;7:259–264. doi: 10.1016/S1471-4914(01)02016-0.
    1. Maggini J, Mirkin G, Bognanni I, Holmberg J, Piazzón IM, Nepomnaschy I, Costa H, Cañones C, Raiden S, Vermeulen M, Geffner JR. Mouse bone marrow-derived mesenchymal stromal cells turn activated macrophages into a regulatory-like profile. PLoS One. 2010;5:e9252. doi: 10.1371/journal.pone.0009252.
    1. Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48:3464–3474. doi: 10.1002/art.11365.
    1. Mizuno H, Tobita M, Uysal AC. Concise review: adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells. 2012;30:804–810. doi: 10.1002/stem.1076.
    1. Erickson GR, Gimble JM, Franklin DM, Rice HE, Awad H, Guilak F. Chondrogenic potential of adipose tissue-derived stromal cells in vitro and in vivo. Biochem Biophys Res Commun. 2002;290:763–769. doi: 10.1006/bbrc.2001.6270.
    1. Guilak F, Lott KE, Awad HA, Cao Q, Hicok KC, Fermor B, Gimble JM. Clonal analysis of the differentiation potential of human adipose-derived adult stem cells. J Cell Physiol. 2006;206:229–237. doi: 10.1002/jcp.20463.
    1. Estes BT, Wu AW, Guilak F. Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis Rheum. 2006;54:1222–1232. doi: 10.1002/art.21779.
    1. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–228. doi: 10.1089/107632701300062859.
    1. Kim BS, Kang KS, Kang SK. Soluble factors from ASCs effectively direct control of chondrogenic fate. Cell Prolif. 2010;43:249–261. doi: 10.1111/j.1365-2184.2010.00680.x.
    1. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109:1292–1298. doi: 10.1161/01.CIR.0000121425.42966.F1.
    1. Kilroy GE, Foster SJ, Wu X, Ruiz J, Sherwood S, Heifetz A, Ludlow JW, Stricker DM, Potiny S, Green P, Halvorsen YD, Cheatham B, Storms RW, Gimble JM. Cytokine profile of human adipose-derived stem cells: expression of angiogenic, hematopoietic, and pro-inflammatory factors. J Cell Physiol. 2007;212:702–709. doi: 10.1002/jcp.21068.
    1. Puissant B, Barreau C, Bourin P, Clavel C, Corre J, Bousquet C, Taureau C, Cousin B, Abbal M, Laharrague P, Penicaud L, Casteilla L, Blancher A. Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells. Br J Haematol. 2005;129:118–129. doi: 10.1111/j.1365-2141.2005.05409.x.
    1. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R, Moseley A, Hoffman R. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002;30:42–48. doi: 10.1016/S0301-472X(01)00769-X.
    1. Pak J. Regeneration of human bones in hip osteonecrosis and human cartilage in knee osteoarthritis with autologous adipose-tissue-derived stem cells: a case series. J Med Case Rep. 2011;5:296. doi: 10.1186/1752-1947-5-296.
    1. Toghraie FS, Chenari N, Gholipour MA, Faghih Z, Torabinejad S, Dehghani S, Ghaderi A. Treatment of osteoarthritis with infrapatellar fat pad derived mesenchymal stem cells in rabbit. Knee. 2011;18:71–75. doi: 10.1016/j.knee.2010.03.001.
    1. Guercio A, Di Marco P, Casella S, Cannella V, Russotto L, Purpari G, Di Bella S, Piccione G. Production of canine mesenchymal stem cells from adipose tissue and their application in dogs with chronic osteoarthritis of the humeroradial joints. Cell Biol Int. 2012;36:189–194. doi: 10.1042/CBI20110304.
    1. Lee JM, Im GI. SOX trio-co-transduced adipose stem cells in fibrin gel to enhance cartilage repair and delay the progression of osteoarthritis in the rat. Biomaterials. 2012;33:2016–2024. doi: 10.1016/j.biomaterials.2011.11.050.
    1. Ter Huurne M, Schelbergen R, Blattes R, Blom A, de Munter W, Grevers L, Jeanson J, Noel D, Casteilla L, Jorgensen C, van den Berg W, van Lent P. Antiinflammatory and chondroprotective effects of intraarticular injection of adipose-derived stem cells in experimental osteoarthritis. Arthritis Rheum. 2012;64:3604–3613. doi: 10.1002/art.34626.
    1. Vignon E, Bejui J, Mathieu P, Hartmann JD, Ville G, Evreux JC, Descotes J. Histological cartilage changes in a rabbit model of osteoarthritis. J Rheumatol. 1987;14(Spec No):104–106.
    1. Grigolo B, Lisignoli G, Desando G, Cavallo C, Marconi E, Tschon M, Giavaresi G, Fini M, Giardino R, Facchini A. Osteoarthritis treated with mesenchymal stem cells on hyaluronan-based scaffold in rabbit. Tissue Eng Part C Methods. 2009;15:647–658. doi: 10.1089/ten.tec.2008.0569.
    1. Fekete N, Gadelorge M, Furst D, Maurer C, Dausend J, Fleury-Cappellesso S, Mailander V, Lotfi R, Ignatius A, Sensebe L, Bourin P, Schrezenmeier H, Rojewski MT. Platelet lysate from whole blood-derived pooled platelet concentrates and apheresis-derived platelet concentrates for the isolation and expansion of human bone marrow mesenchymal stromal cells: production process, content and identification of active components. Cytotherapy. 2012;14:540–554. doi: 10.3109/14653249.2012.655420.
    1. Valorani MG, Montelatici E, Germani A, Biddle A, D'Alessandro D, Strollo R, Patrizi MP, Lazzari L, Nye E, Otto WR, Pozzilli P, Alison MR. Pre-culturing human adipose tissue mesenchymal stem cells under hypoxia increases their adipogenic and osteogenic differentiation potentials. Cell Prolif. 2012;45:225–238. doi: 10.1111/j.1365-2184.2012.00817.x.
    1. Weir C, Morel-Kopp MC, Gill A, Tinworth K, Ladd L, Hunyor SN, Ward C. Mesenchymal stem cells: isolation, characterisation and in vivo fluorescent dye tracking. Heart Lung Circ. 2008;17:395–403. doi: 10.1016/j.hlc.2008.01.006.
    1. Bouchgua M, Alexander K, d'Anjou MA, Girard CA, Carmel EN, Beauchamp G, Richard H, Laverty S. Use of routine clinical multimodality imaging in a rabbit model of osteoarthritis--part I. Osteoarthritis Cartilage. 2009;17:188–196. doi: 10.1016/j.joca.2008.06.017.
    1. Papaioannou NA, Triantafillopoulos IK, Khaldi L, Krallis N, Galanos A, Lyritis GP. Effect of calcitonin in early and late stages of experimentally induced osteoarthritis. A histomorphometric study. Osteoarthritis Cartilage. 2007;15:386–395. doi: 10.1016/j.joca.2006.10.012.
    1. Pastoureau P, Leduc S, Chomel A, De Ceuninck F. Quantitative assessment of articular cartilage and subchondral bone histology in the meniscectomized guinea pig model of osteoarthritis. Osteoarthritis Cartilage. 2003;11:412–423. doi: 10.1016/S1063-4584(03)00050-5.
    1. Laverty S, Girard CA, Williams JM, Hunziker EB, Pritzker KP. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the rabbit. Osteoarthritis Cartilage. 2010;18(Suppl 3):S53–S65.
    1. Desando G, Cavallo C, Tschon M, Giavaresi G, Martini L, Fini M, Giardino R, Facchini A, Grigolo B. Early-term effect of adult chondrocyte transplantation in an osteoarthritis animal model. Tissue Eng Part A. 2012;18:1617–1627. doi: 10.1089/ten.tea.2011.0494.
    1. Schäffler A, Büchler C. Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25:818–827. doi: 10.1634/stemcells.2006-0589.
    1. Nathan S, Das De S, Thambyah A, Fen C, Goh J, Lee EH. Cell-based therapy in the repair of osteochondral defects: a novel use for adipose tissue. Tissue Eng. 2003;9:733–744. doi: 10.1089/107632703768247412.
    1. Mokbel AN, El Tookhy OS, Shamaa AA, Rashed LA, Sabry D, El Sayed AM. Homing and reparative effect of intra-articular injection of autologus mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord. 2011;12:259. doi: 10.1186/1471-2474-12-259.
    1. Lefebvre V, Peeters-Joris C, Vaes G. Modulation by interleukin 1 and tumor necrosis factor alpha of production of collagenase, tissue inhibitor of metalloproteinases and collagen types in differentiated and dedifferentiated articular chondrocytes. Biochim Biophys Acta. 1990;1052:366–378. doi: 10.1016/0167-4889(90)90145-4.
    1. Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7:33–42. doi: 10.1038/nrrheum.2010.196.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    1. Le Blanc K, Mougiakakos D. Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol. 2012;12:383–396. doi: 10.1038/nri3209.
    1. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, Robey PG, Leelahavanichkul K, Koller BH, Brown JM, Hu X, Jelinek I, Star RA, Mezey E. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–49. doi: 10.1038/nm.1905.
    1. Crop MJ, Baan CC, Korevaar SS, Ijzermans JN, Pescatori M, Stubbs AP, van Ijcken WF, Dahlke MH, Eggenhofer E, Weimar W, Hoogduijn MJ. Inflammatory conditions affect gene expression and function of human adipose tissue-derived mesenchymal stem cells. Clin Exp Immunol. 2010;162:474–486. doi: 10.1111/j.1365-2249.2010.04256.x.
    1. Goldring MB, Otero M, Plumb DA, Dragomir C, Favero M, El Hachem K, Hashimoto K, Roach HI, Olivotto E, Borzì RM, Marcu KB. Roles of inflammatory and anabolic cytokines in cartilage metabolism: signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur Cell Mater. 2011;21:202–220.
    1. Kim WS, Park BS, Sung JH. The wound-healing and antioxidant effects of adipose-derived stem cells. Expert Opin Biol Ther. 2009;9:879–887. doi: 10.1517/14712590903039684.

Source: PubMed

3
Subscribe