Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue and its derived mesenchymal stromal cells

Valentina Ceserani, Anna Ferri, Angiola Berenzi, Anna Benetti, Emilio Ciusani, Luisa Pascucci, Cinzia Bazzucchi, Valentina Coccè, Arianna Bonomi, Augusto Pessina, Erica Ghezzi, Offer Zeira, Piero Ceccarelli, Silvia Versari, Carlo Tremolada, Giulio Alessandri, Valentina Ceserani, Anna Ferri, Angiola Berenzi, Anna Benetti, Emilio Ciusani, Luisa Pascucci, Cinzia Bazzucchi, Valentina Coccè, Arianna Bonomi, Augusto Pessina, Erica Ghezzi, Offer Zeira, Piero Ceccarelli, Silvia Versari, Carlo Tremolada, Giulio Alessandri

Abstract

Background: Adipose-derived mesenchymal stromal cells (Ad-MSCs) are a promising tool for advanced cell-based therapies. They are routinely obtained enzymatically from fat lipoaspirate (LP) as SVF, and may undergo prolonged ex vivo expansion, with significant senescence and decline in multipotency. Besides, these techniques have complex regulatory issues, thus incurring in the compelling requirements of GMP guidelines. Hence, availability of a minimally manipulated, autologous adipose tissue would have remarkable biomedical and clinical relevance. For this reason, a new device, named Lipogems® (LG), has been developed. This ready-to-use adipose tissue cell derivate has been shown to have in vivo efficacy upon transplantation for ischemic and inflammatory diseases. To broaden our knowledge, we here investigated the angiogenic and anti-inflammatory properties of LG and its derived MSC (LG-MSCs) population.

Methods: Human LG samples and their LG-MSCs were analyzed by immunohistochemistry for pericyte, endothelial and mesenchymal stromal cell marker expression. Angiogenesis was investigated testing the conditioned media (CM) of LG (LG-CM) and LG-MSCs (LG-MSCs-CM) on cultured endothelial cells (HUVECs), evaluating proliferation, cord formation, and the expression of the adhesion molecules (AM) VCAM-1 and ICAM-1. The macrophage cell line U937 was used to evaluate the anti-inflammatory properties, such as migration, adhesion on HUVECs, and release of RANTES and MCP-1.

Results: Our results indicate that LG contained a very high number of mesenchymal cells expressing NG2 and CD146 (both pericyte markers) together with an abundant microvascular endothelial cell (mEC) population. Substantially, both LG-CM and LG-MSC-CM increased cord formation, inhibited endothelial ICAM-1 and VCAM-1 expression following TNFα stimulation, and slightly improved HUVEC proliferation. The addition of LG-CM and LG-MSC-CM strongly inhibited U937 migration upon stimulation with the chemokine MCP-1, reduced their adhesion on HUVECs and significantly suppressed the release of RANTES and MCP-1.

Conclusions: Our data indicate that LG micro-fragmented adipose tissue retains either per se, or in its embedded MSCs content, the capacity to induce vascular stabilization and to inhibit several macrophage functions involved in inflammation.

Figures

Fig. 1
Fig. 1
Histological and immunohistochemical analysis of LG and LP. H&E staining shows higher content of microvessels in LG (a) compared to LP (b). The immunohistochemistry shows an intense staining (black arrows) for the endothelial marker CD31 (c and d) and mural cell marker SMA (e and f). Staining with NG2 (g and h) was associated to microvessels and showed the presence of pericytes, that were more abundant in LG compared to LP (black arrows). Staining for CD146 (i and j), a marker present on both pericytes and endothelial cells, was more diffuse and intense in LP, while in LG was more associated to microvessels (black arrows). VEGF staining was present in the connective of both LG (k) and LP (l); LG showed less connective matrix than LP (Magnification 20x)
Fig. 2
Fig. 2
Cultures of LG-MSCs upon collagenase dissociation. Representative of five different Lipogems®-derived MSCs (LG-MSC) cultured from 1 h (a) to 21 days (f). Upon enzymatic digestion of human LG specimens, the obtained clusters were seeded in IMDM + 5 % FBS (a) in order to allow the adhesion and the proliferation of the released cells. After 72 h (b) all cultures showed the capacity to adhere on a plastic substrate and the typical stromal morphology. Starting from 5 days of culture (c) clusters of endothelial cells (black arrow) were highly distinguishable among the stromal cells and after 10 days (d) were removed from the culture by using CD31+ magnetic beads. Stromal and endothelial cell population was cultured separately and at 21 days reached complete confluence (e) and (f) respectively. (Magnification 10x)
Fig. 3
Fig. 3
Immunostaining of early culture of cells obtained after collagenase digestion of LG. Endothelial, pericyte and mesenchymal markers were used to characterize the cells extracted from LG. Cells analyzed upon 48-72 h of culture, showed the presence of a significant number of cell colonies positive for CD31, CD34 endothelial markers, surrounded by fibroblastic negative cells (black arrows). Conversely, several fibroblastic-like cells were positive for the pericyte marker NG2, and the mesenchymal markers CD105, CD44 and SMA. Interestingly, CD146 stained very intensely both colonies and fibroblastic cells; very few cells were negative. Fibroblastic-like cells were also positive for TGFβ1 and VEGF, growth factors that are usually secreted by MSCs. (Magnification 20x)
Fig. 4
Fig. 4
Immunocytochemistry of secondary culture of LG-derived MSCs. Immunocytochemistry was performed on LG-MSCs after 2–3 in vitro passages. Under these experimental conditions, LG-MSCs lost the expression of endothelial markers CD31, CD34 and KDR (a receptor for VEGF), while retaining pericyte and mesenchymal markers NG2, CD105 and SMA. Cells were still positive for TGFβ1 and VEGF. To note, CD146 positive cells (black arrow) were very few, suggesting that this marker in LG was probably mostly expressed by the cells of endothelial origin (Magnification 20x)
Fig. 5
Fig. 5
Characterization of Lipogems®-derived MSCs. The levels of CD44-PE, CD73-PE, CD90-PE, CD105-PE, CD31-PE markers in LG-MSCs were analyzed by flow cytometry. The percentage indicate the positivity of the cells for their relative surface markers, while the gate indicate the isotypic control (Magnification 10x)
Fig. 6
Fig. 6
Angiogenic properties of LG and LG-MSCs. The angiogenic properties of LG and LG-MSCs were investigated by testing their CM. In (a) the effect of LG-CM and LG-MSCs-CM on HUVECs proliferation. EGM medium was used as positive control for proliferation, EGM medium without growth factors the negative control (CTRL) (* p < 0.05; ** p < 0.01 versus CTRL). In (b) the quantification of cord formations on MTG by HUVECs under the influence of LG-CM and LG-MSCs-CM at 12 h and at 24 h in (c) (* p < 0.05; ** p < 0.01 versus CTRL). In (d) the analysis of angiogenic molecules present in the secretome of LG-CM in comparison to LG-MSCs-CM. MMP2 was significantly reduced in LG-CM, while ANG-1 and ANG-2 were increased (* p < 0.05; ** p < 0.01 versus LG-MSCs-CM). In (e) and in (f) the effect of LG-CM and LG-MSCs-CM on the expression of HUVEC ICAM-1 and VCAM-1 respectively (* p < 0.05; ** p < 0.01 versus CTRL; °p < 0.05, °° p < 0.01 versus TNFα treatment)
Fig. 7
Fig. 7
LG-CM and LG-MSCs possess anti-inflammatory properties. The capacity of LG-CM and LG-MSCs to affect inflammatory cell functions was tested on the U937 monocyte cell line. In (a) the effect of LG-CM and LG-MSCs-CM on U937 migration. Different dilutions of LG-CM and LG-MSCs-CM were placed in the lower compartment of a transwell insert (5um pore size). The chemokine MCP-1 at (10 ng/ml) was used as a positive stimuli for U937 migration. RPMI+ 0.2 % BSA was the control medium (CTRL) (* p < 0.05; ** p < 0.01 versus CTRL; °p < 0.05, versus MCP-1 treatment). In (b) adhesion of U937 to HUVECs monolayer in the presence of LG-CM and LG-MSCs-CM. TNFα at 20 ng/ml was used to activate HUVECs (* p < 0.05; ** p < 0.01 versus CTRL; °p < 0.05, °° p < 0.01 versus TNFα treatment). The photos below the figure show the morphological appearance of U937 attached to HUVECs primed with TNFα, LG-CM and LG-MSCs-CM after 30’ incubation (5x magnifications). In (c) and in (d) the release of RANTES and MCP-1 by U937 under the influence of LG-CM and LG-MSCs-CM stimulated or not with LPS (1ug/ml) respectively (*p < 0.05; **p < 0.01 versus CTRL; °p < 0.05, °° p < 0.01 versus LPS treatment)

References

    1. Alessandri G, Chirivi RG, Fiorentini S, Dossi R, Bonardelli S, Giulini SM, Zanetta G, Landoni F, Graziotti PP, Turano A, Caruso A, Zardi L, Giavazzi R, Bani MR. Phenotypic and functional characteristics of tumour-derived microvascular endothelial cells. Clin Exp Metastasis. 1999;17:655–662. doi: 10.1023/A:1006738901839.
    1. Bardin N, Reumaux D, Geboes K, Colombel JF, Blot-Chabaud M, Sampol J, Duthilleul P, Dignat-George F. Increased expression of CD146, a new marker of the endothelial junction in active inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:16–21. doi: 10.1097/01.MIB.0000194181.46930.88.
    1. Benetti A, Berenzi A, Gambarotti M, Garrafa E, Gelati M, Dessy E, Portolani N, Piardi T, Giulini SM, Caruso A, Invernici G, Parati EA, Nicosia R, Alessandri G. Transforming growth factor-beta1 and CD105 promote the migration of hepatocellular carcinoma-derived endothelium. Cancer Res. 2008;68:8626–34. doi: 10.1158/0008-5472.CAN-08-1218.
    1. Bianchi F, Maioli M, Leonardi E, Olivi E, Pasquinelli G, Valente S, Mendez AJ, Ricordi C, Raffaini M, Tremolada C, Ventura C. A new nonenzymatic method and device to obtain a fat tissue derivative highly enriched in pericyte-like elements by mild mechanical forces from human lipoaspirates. Cell Transplant. 2013;22(11):2063–77. doi: 10.3727/096368912X657855.
    1. Bianchi F, Olivi M, Baldassarre M, Giannone FA, Laggetta M, Valente S, Cavallini C, Tassinari R, Canaider S, Pasquinelli G, Tremolada C, Ventura C. Lipogems, a New Modality of Fat Tissue Handling to Enhance Tissue Repair in Chronic Hind Limb Ischemia. CellR4. 2014;2(6):e1289.
    1. Blasi A, Martino C, Balducci L, Saldarelli M, Soleti A, Navone SE, Canzi L, Cristini S, Invernici G, Parati EA, Alessandri G. Dermal fibroblasts display similar phenotypic and differentiation capacity to fat-derived mesenchymal stem cells, but differ in anti-inflammatory and angiogenic potential. Vasc Cell. 2011;3(1):5. doi: 10.1186/2045-824X-3-5.
    1. Caplan AI. All MSCs are pericytes? Cell Stem Cell. 2008;3(3):229–30. doi: 10.1016/j.stem.2008.08.008.
    1. Caplan AI. Why are MSCs therapeutic? New data: new insight. J Pathol. 2009;217:318–324. doi: 10.1002/path.2469.
    1. Carelli S, Messaggio F, Canazza A, Hebda DM, Caremoli F, Latorre E, Grimoldi MG, Colli M, Bulfamante G, Tremolada C, Di Giulio AM, Gorio A. Characteristics and Properties of Mesenchymal Stem Cells Derived From Microfragmented Adipose Tissue. Cell Transplant. 2015;24(7):1233–52. doi: 10.3727/096368914X681603.
    1. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, Norotte C, Teng PN, Traas J, Schugar R, Deasy BM, Badylak S, Buhring HJ, Giacobino JP, Lazzari L, Huard J, Péault B. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3(3):301–13. doi: 10.1016/j.stem.2008.07.003.
    1. De Siena R, Balducci L, Blasi A, Montanaro MG, Saldarelli M, Saponaro V, Martino C, Logrieco G, Soleti A, Fiobellot S, Madeddu P, Rossi G, Ribatti D, Crovace A, Cristini S, Invernici G, Parati EA, Alessandri G. Omentum-derived stromal cells improve myocardial regeneration in pig post-infarcted heart through a potent paracrine mechanism. Exp Cell Res. 2010;316:1804–1815. doi: 10.1016/j.yexcr.2010.02.009.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop DJ, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7. doi: 10.1080/14653240600855905.
    1. Fagiani E, Christofori G. Angiopoietins in angiogenesis. Cancer Lett. 2013;328:18–26. doi: 10.1016/j.canlet.2012.08.018.
    1. García-Contreras M, Messaggio F, Jimenez O, Mendez A. Differences in exosome content of human adipose tissue processed by non-enzymatic and enzymatic methods. CellR4. 2015;3(1):e1423.
    1. Garrafa E, Alessandri G, Benetti A, Turetta D, Corradi A, Cantoni AM, Cervi E, Bonardelli S, Parati E, Giulini SM, Ensoli B, Caruso A. Isolation and characterization of lymphatic microvascular endothelial cells from human tonsils. J Cell Physiol. 2006;207:107–13. doi: 10.1002/jcp.20537.
    1. Hall AP. Review of the pericyte during angiogenesis and its role in cancer and diabetic retinopathy. Toxicol Pathol. 2006;34(6):763–75. doi: 10.1080/01926230600936290.
    1. Hao H, Liu J, Shen J, Zhao Y, Liu H, Hou Q, Tong C, Ti D, Dong L, Cheng Y, Mu Y, Liu J, Fu X, Han W. Multiple intravenous infusions of bone marrow mesenchymal stem cells reverse hyperglycemia in experimental type 2 diabetes rats. Biochem Biophys Res Commun. 2013;436:418–23. doi: 10.1016/j.bbrc.2013.05.117.
    1. Hirschi KK, D'Amore PA. Pericytes in the microvasculature. Cardiovasc Res. 1996;32:687–98. doi: 10.1016/S0008-6363(96)00063-6.
    1. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol. 2005;15:378–386. doi: 10.1016/j.semcancer.2005.05.004.
    1. Langer HF, Chavakis T. Leukocyte-endothelial interactions in inflammation. J Cell Mol Med. 2009;13(7):1211–20. doi: 10.1111/j.1582-4934.2009.00811.x.
    1. Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, Bae YC, Jung JS. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 2004;14(4–6):311–324. doi: 10.1159/000080341.
    1. Lee RH, Yu JM, Foskett AM, Peltier G, Reneau JC, Bazhanov N, Oh JY, Prockop DJ. TSG-6 as a biomarker to predict efficacy of human mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation in vivo. PNAS. 2014;111(47):16766–71. doi: 10.1073/pnas.1416121111.
    1. Lehmann MH. Recombinant human granulocyte-macrophage colony-stimulating factor triggers interleukin-10 expression in the monocytic cell line U937. Mol Immunol. 1998;35(8):479–485. doi: 10.1016/S0161-5890(98)00043-1.
    1. Liu WH, Song FQ, Ren LN, Guo WQ, Wang T, Feng YX, Tang LJ, Li K. The multiple functional roles of mesenchymal stem cells in participating in treating liver diseases. J Cell Mol Med. 2015;19:511–20. doi: 10.1111/jcmm.12482.
    1. Lloyd CM, Dorf ME, Proudfoot A, Salant DJ, Gutierrez-Ramos JC. Role of MCP-1 and RANTES in inflammation and progression to fibrosis during murine crescentic nephritis. J Leukoc Biol. 1997;62(5):676–80.
    1. McHale JF, Harari OA, Marshall D, Haskard DO. Vascular Endothelial Cell Expression of ICAM-1 and VCAM-1 at the Onset of Eliciting Contact Hypersensitivity in Mice: Evidence for a Dominant Role of TNF-alpha. J Immunol. 1999;162:1648–1655.
    1. Merfeld-Clauss S, Li J, Kolonin M, Arap W, Pasqualini R, Johnstone BH, March KL. A populationof multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers reside in a periendothelial location, and stabilize endothelial networks. Circ Res. 2008;102:77–85. doi: 10.1161/CIRCRESAHA.107.159475.
    1. Mosna F, Sensebé L, Krampera M. Human Bone-Marrow And Adipose Tissue Mesenchymal Stem Cells: A User's Guide. Stem Cells Dev. 2010;19:1449–70. doi: 10.1089/scd.2010.0140.
    1. Muller WA, Randolph GJ. Migration of leukocytes across endothelium and beyond: molecules involved in the transmigration and fate of monocytes. J Leukoc Biol. 1999;66(5):698–704.
    1. Park KS, Kim YS, Kim JH, Choi B, Kim SH, Tan AH, Lee MS, Lee MK, Kwon CH, Joh JW, Kim SJ, Kim KW. Trophic molecules derived from human mesenchymal stem cells enhance survival, function, and angiogenesis of isolated islets after transplantation. Transplantation. 2010;89:509–517.
    1. Pessina A, Coccè V, Pascucci L, Bonomi A, Cavicchini L, Sisto F, Ferrari M, Ciusani E, Crovace A, Falchetti ML, Zicari S, Caruso A, Navone S, Marfia G, Benetti A, Ceccarelli P, Parati E, Alessandri G. Mesenchymal stromal cells primed with Paclitaxel attract and kill leukaemia cells, inhibit angiogenesis and improve survival of leukaemia-bearing mice. Br J Haematol. 2013;160:766–78. doi: 10.1111/bjh.12196.
    1. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143.
    1. Plock, J.A.; Schnider, J.T.; Zhang, W.; Schweizer, R.; Tsuji, W.; Kostereva, N.; Fanzio, P.M.; Ravuri, S.; Solari, M.G.; Cheng, H.Y.; Rubin, P.J.; Marra, K.G.; Gorantla, V.S. Adipose - and Bone Marrow-Derived Mesenchymal Stem Cells Prolong Graft Survival in Vascularized Composite Allotransplantation. Transplantation. Jun 22; 2015 [Epub ahead of print].
    1. Prockop DJ. Concise review: two negative feedback loops place mesenchymal stem/stromal cells at the center of early regulators of inflammation. Stem Cells. 2013;31(10):2042–6. doi: 10.1002/stem.1400.
    1. Raffaini M, Tremolada C. Micro Fractured and Purified Adipose Tissue Graft (Lipogems®) Can Improve the Orthognathic Surgery Outcomes Both Aesthetically and in Postoperative Healing. CellR4. 2014;2(4):e1118.
    1. Roseti L, Serra M, Tigani D, Brognara I, Lopriore A, Bassi A, Fornasari PM. Cell manipulation in autologous chondrocyte implantation: from research to cleanroom. Chir Organi Mov. 2008;91:147–51. doi: 10.1007/s12306-007-0024-1.
    1. Suk K, Cha S. Thrombin-induced interleukin-8 production and its regulation by interferon-gamma and prostaglandin E2 in human monocytic U937 cells. Immunol Lett. 1999;67(3):223–27. doi: 10.1016/S0165-2478(99)00015-2.
    1. Tremolada C, Palmieri G, Ricordi C. Adipocyte transplantation and stem cells: plastic surgery meets regenerative medicine. Cell Transplant. 2010;19:1217–1223. doi: 10.3727/096368910X507187.
    1. Tremolada C, Beltrami G, Magri A. Adipose mesenchymal stem cells and “Regenerative adipose tissue graft” (LIPOGEMSTM) for musculoskeletal regeneration. Eur J Musculoskelet Dis. 2014;3(2):57–67.
    1. Xu LL, Warren MK, Rose WL, Gong W, Wang JM. Human recombinant monocyte chemotactic protein and other C-C chemokines bind and induce directional migration of dendritic cells in vitro. J Leukoc Biol. 1996;60(3):365–371.
    1. Zhu Y, Song X, Han F, Li Y, Wei J, Liu X. Alteration of histone acetylation pattern during long-term serum-free culture conditions of human fetal placental mesenchymal stem cells. PLoS One. 2015;10(2):e0117068. doi: 10.1371/journal.pone.0117068.
    1. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105.

Source: PubMed

3
Sottoscrivi