Vasculogenic and angiogenic potential of adipose stromal vascular fraction cell populations in vitro

Joseph S Zakhari, Jacob Zabonick, Brian Gettler, Stuart K Williams, Joseph S Zakhari, Jacob Zabonick, Brian Gettler, Stuart K Williams

Abstract

Adipose-derived stromal vascular fraction (SVF) is a heterogeneous cell source that contains endothelial cells, pericytes, smooth muscle cells, stem cells, and other accessory immune and stromal cells. The SVF cell population has been shown to support vasculogenesis in vitro as well vascular maturation in vivo. Matrigel, an extracellular matrix (ECM) mixture has been utilized in vitro to evaluate tube formation of purified endothelial cell systems. We have developed an in vitro system that utilizes freshly isolated SVF and ECM molecules both in pure form (fibrin, laminin, collagen) as well as premixed form (Matrigel) to evaluate endothelial tip cell formation, endothelial stalk elongation, and early stages of branching and inosculation. Freshly isolated SVF rat demonstrate cell aggregation and clustering (presumptive vasculogenesis) on Matrigel ECM within the first 36 h of seeding followed by tip cell formation, stalk cell formation, branching, and inosculation (presumptive angiogenesis) during the subsequent 4 days of culture. Purified ECM molecules (laminin, fibrin, and collagen) promote cell proliferation but do not recapitulate events seen on Matrigel. We have created an in vitro system that provides a functional assay to study the mechanisms of vasculogenesis and angiogenesis in freshly isolated SVF to characterize SVF's blood vessel forming potential prior to clinical implantation.

Keywords: Adipose; Angiogenesis; Stromal vascular fraction; Vasculogenesis.

Figures

Figure 1.
Figure 1.
×4 phase contrast images of SVF grown on 3 mg/mL collagen I (A), 1% gelatin (B), Matrigel (C), 5 mg/mL fibrin (D), or 2 μg laminin 332 (E) after 112 h of incubation. Monolayers of cells are formed on collagen I (A), gelatin (B) and laminin 332 (E) with cell clustering demonstrated on Matrigel (C) and fibrin (D). SVF forms microvascular networks on Matrigel (C).
Figure 2.
Figure 2.
×4 phase contrast images of SVF grown on Matrigel at 18 h (A, B); 36 h (C, D); 60 h (E, F); and 112 h (G, H). Images B, D, F, and H are ×3 optical zooms of boxed sections of images A, C, E, and G, respectively. White arrows demonstrate initial cell clustering occurring at 18 h. Tip cell formation occurs at 36 h as marked by the red arrow. Stalk cell formation and elongation occurs at 60 h as marked by the blue arrow. Branching (green arrow) and inosculation (yellow arrow) can be seen at 118 h in vitro.
Figure 3.
Figure 3.
×10 fluorescent images of SVF grown on 1% gelatin (A) or Matrigel (B) for 112 h. Blue: DAPI, green: Griffonia simplicifolia 1 endothelial specific lectin-FITC, and red: α smooth muscle actin-rhodamine.
Figure 4.
Figure 4.
Schematic of SVF microvascular assembly on Matrigel in vitro. Digested SVF cells (A) self-assemble into a cluster of cells by 18 h (B). By 36 h, endothelial cells sprout out of the cluster with dendritic-like extensions (tip cells: orange ●) (C). DAPT, an inhibitor of the γ-secretase that cleaves notch to its soluble ligand, blocks tip cell signaling to stalk cells, inhibiting stalk cell formation. Stalk cell formation occurs when notch signaling leads to a higher expression of VEGR-1 on the adjacent cell. ZM 306416 blocks VEGFR-1 and thus, stalk cell phenotype. In the absence of inhibitors, stalk cells (black ) migrate away from clusters behind tip cells by 60 h (D). Microvascular networks continue to grow after 112 h and demonstrate increased complexity (E). Networks from adjacent clusters inosculate with one another (yellow *) and recruit perivascular support cells to endothelial tubes (purple ▲) (F). PDGFR-B is found on perivascular support cells and activated by endothelial PDGF-β, which signals pericyte recruitment and tube stabilization. Imatinib mesylate inhibits PDGFR-B.
Figure 5.
Figure 5.
Number of SVF cell clusters (grouping of two or more cells over 25 μm in total size) at 18, 36, 60, and 112 h of incubation in the presence of inhibitors (A). Cluster size (micrometer) at 18, 36, 60, and 112 h of incubation in the presence of inhibitors (B). Percentage of clusters containing tip cells at 18, 36, 60, and 112 h of incubation in the presence of inhibitors (C). Percentage of clusters containing stalk cells at 18, 36, 60, and 112 h of incubation in the presence of inhibitors (D). Angiogenic inhibitors (in DMSO) were added at days 0, 2, 4, and 6. p < 0.05 *, p < 0.005 **, p < 0.0005 ***.

References

    1. Albini A. Extracellular matrix invasion in metastases and angiogenesis: commentary on the Matrigel “Chemoinvasion assay”. Cancer Res. 2016;76(16):4595–4597. doi: 10.1158/0008-5472.CAN-16-1971.
    1. Antczak C, et al. A high-content biosensor-based screen identifies cell-permeable activators and inhibitors of EGFR function: implications in drug discovery. J Biomol Screen. 2012;17(7):885–899. doi: 10.1177/1087057112446174.
    1. Arnaoutova I, Kleinman HK. In vitro angiogenesis: endothelial cell tube formation on gelled basement membrane extract. Nat Protoc. 2010;5(4):628–635. doi: 10.1038/nprot.2010.6.
    1. Blanco R, Gerhardt H. VEGF and notch in tip and stalk cell selection. Cold Spring Harb Perspect Med. 2013;3(1):a006569. doi: 10.1101/cshperspect.a006569.
    1. Bogorad MI et al (2017) Tissue-engineered 3D microvessel and capillary network models for the study of vascular phenomena. Microcirculation
    1. Brown RM, et al. Tube-forming assays. Methods Mol Biol. 2016;1430:149–157. doi: 10.1007/978-1-4939-3628-1_9.
    1. Carstens MH, et al. Non-reconstructable peripheral vascular disease of the lower extremity in ten patients treated with adipose-derived stromal vascular fraction cells. Stem Cell Res. 2017;18:14–21. doi: 10.1016/j.scr.2016.12.001.
    1. Chang CC, et al. Determinants of microvascular network topologies in implanted neovasculatures. Arterioscler Thromb Vasc Biol. 2012;32(1):5–14. doi: 10.1161/ATVBAHA.111.238725.
    1. Diaz-Flores L, et al. Behavior of in situ human native adipose tissue CD34+ stromal/progenitor cells during different stages of repair. Tissue-resident CD34+ stromal cells as a source of myofibroblasts. Anat Rec (Hoboken) 2015;298(5):917–930. doi: 10.1002/ar.23086.
    1. Ebrahem Q, et al. Cross-talk between vascular endothelial growth factor and matrix metalloproteinases in the induction of neovascularization in vivo. Am J Pathol. 2010;176(1):496–503. doi: 10.2353/ajpath.2010.080642.
    1. Fercana GR, et al. Perivascular extracellular matrix hydrogels mimic native matrix microarchitecture and promote angiogenesis via basic fibroblast growth factor. Biomaterials. 2017;123:142–154. doi: 10.1016/j.biomaterials.2017.01.037.
    1. Gaengel K, et al. Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler Thromb Vasc Biol. 2009;29(5):630–638. doi: 10.1161/ATVBAHA.107.161521.
    1. Haug V, et al. Comparison between endothelial progenitor cells and human umbilical vein endothelial cells on neovascularization in an adipogenesis mouse model. Microvasc Res. 2015;97:159–166. doi: 10.1016/j.mvr.2014.10.005.
    1. Heymach JV, et al. Phase II study of the antiangiogenic agent SU5416 in patients with advanced soft tissue sarcomas. Clin Cancer Res. 2004;10(17):5732–5740. doi: 10.1158/1078-0432.CCR-04-0157.
    1. Hiscox AM, et al. An islet-stabilizing implant constructed using a preformed vasculature. Tissue Eng Part A. 2008;14(3):433–440. doi: 10.1089/tea.2007.0099.
    1. Hughes SE. Functional characterization of the spontaneously transformed human umbilical vein endothelial cell line ECV304: use in an in vitro model of angiogenesis. Exp Cell Res. 1996;225(1):171–185. doi: 10.1006/excr.1996.0168.
    1. Jin E, et al. Angiogenic characteristics of human stromal vascular fraction in ischemic hindlimb. Int J Cardiol. 2017;234:38–47. doi: 10.1016/j.ijcard.2017.02.080.
    1. Kc P, et al. Prevascularization of decellularized porcine myocardial slice for cardiac tissue engineering. ACS Appl Mater Interfaces. 2017;9(3):2196–2204. doi: 10.1021/acsami.6b15291.
    1. Laschke MW, Menger MD. Adipose tissue-derived microvascular fragments: natural vascularization units for regenerative medicine. Trends Biotechnol. 2015;33(8):442–448. doi: 10.1016/j.tibtech.2015.06.001.
    1. Laschke MW, Menger MD. Prevascularization in tissue engineering: current concepts and future directions. Biotechnol Adv. 2016;34(2):112–121. doi: 10.1016/j.biotechadv.2015.12.004.
    1. Leblanc AJ, Touroo JS, Hoying JB, Williams SK (2012) Adipose stromal vascular fraction cell construct sustains coronary microvascular function after acute myocardial infarction. Am J Physiol Heart Circ Physiol 302(4):H973–H982
    1. Leblanc AJ, Nguyen QT, Touroo JS, Aird AL, Chang RC, Ng CK, Hoying JB, Williams SK (2013) Adipose-derived cell construct stabilizes heart function and increases microvascular perfusion in an established infarct. Stem Cells Transl Med 2(11):896–905
    1. Lin S, et al. Smad signal pathway regulates angiogenesis via endothelial cell in an adipose-derived stromal cell/endothelial cell co-culture, 3D gel model. Mol Cell Biochem. 2016;412(1–2):281–288. doi: 10.1007/s11010-015-2634-5.
    1. Madri JA, Williams SK. Capillary endothelial cell cultures: phenotypic modulation by matrix components. J Cell Biol. 1983;97(1):153–165. doi: 10.1083/jcb.97.1.153.
    1. Maijub JG, et al. Concentration-dependent vascularization of adipose stromal vascular fraction cells. Cell Transplant. 2015;24(10):2029–2039. doi: 10.3727/096368914X685401.
    1. Mammoto T, et al. Extracellular matrix structure and tissue stiffness control postnatal lung development through the lipoprotein receptor-related protein 5/tie2 signaling system. Am J Respir Cell Mol Biol. 2013;49(6):1009–1018. doi: 10.1165/rcmb.2013-0147OC.
    1. Mildmay-White A, Khan W (2016) Cell surface markers on adipose-derived stem cells: a systematic review. Curr Stem Cell Res Ther
    1. Moore MC, Pandolfi V, McFetridge PS. Novel human-derived extracellular matrix induces in vitro and in vivo vascularization and inhibits fibrosis. Biomaterials. 2015;49:37–46. doi: 10.1016/j.biomaterials.2015.01.022.
    1. Morrissette-McAlmon J, et al. J Tissue Eng Regen Med. 2017. Adipose-derived perivascular mesenchymal stromal/stem cells promote functional vascular tissue engineering for cardiac regenerative purposes.
    1. Nicosia RF, Ottinetti A. Modulation of microvascular growth and morphogenesis by reconstituted basement membrane gel in three-dimensional cultures of rat aorta: a comparative study of angiogenesis in Matrigel, collagen, fibrin, and plasma clot. In Vitro Cell Dev Biol. 1990;26(2):119–128. doi: 10.1007/BF02624102.
    1. Nourse MB et al (2009) VEGF induces differentiation of functional endothelium from human embryonic stem cells. Implications for tissue engineering. Arterioscler Thromb Vasc Biol
    1. Nunes SS, et al. Generation of a functional liver tissue mimic using adipose stromal vascular fraction cell-derived vasculatures. Sci Rep. 2013;3:2141. doi: 10.1038/srep02141.
    1. Riemenschneider SB, et al. Inosculation and perfusion of pre-vascularized tissue patches containing aligned human microvessels after myocardial infarction. Biomaterials. 2016;97:51–61. doi: 10.1016/j.biomaterials.2016.04.031.
    1. Sacharidou A, Stratman AN, Davis GE. Molecular mechanisms controlling vascular lumen formation in three-dimensional extracellular matrices. Cells Tissues Organs. 2012;195(1–2):122–143. doi: 10.1159/000331410.
    1. Sasagawa T, et al. Endothelial colony-forming cells for preparing prevascular three-dimensional cell-dense tissues using cell-sheet engineering. J Tissue Eng Regen Med. 2016;10(9):739–747. doi: 10.1002/term.1858.
    1. Shepherd BR, Hoying JB, Williams SK. Microvascular transplantation after acute myocardial infarction. Tissue Eng. 2007;13(12):2871–2879. doi: 10.1089/ten.2007.0025.
    1. Staton CA, Reed MW, Brown NJ. A critical analysis of current in vitro and in vivo angiogenesis assays. Int J Exp Pathol. 2009;90(3):195–221. doi: 10.1111/j.1365-2613.2008.00633.x.
    1. Sun X, Altalhi W, Nunes SS. Vascularization strategies of engineered tissues and their application in cardiac regeneration. Adv Drug Deliv Rev. 2016;96:183–194. doi: 10.1016/j.addr.2015.06.001.
    1. Tung JJ, Tattersall IW, Kitajewski J. Tips, stalks, tubes: notch-mediated cell fate determination and mechanisms of tubulogenesis during angiogenesis. Cold Spring Harb Perspect Med. 2012;2(2):a006601. doi: 10.1101/cshperspect.a006601.
    1. Valarmathi MT, et al. A novel human tissue-engineered 3-D functional vascularized cardiac muscle construct. Front Cell Dev Biol. 2017;5:2. doi: 10.3389/fcell.2017.00002.
    1. Wagner RC, Matthews MA. The isolation and culture of capillary endothelium from epididymal fat. Microvasc Res. 1975;10(3):286–297. doi: 10.1016/0026-2862(75)90033-3.
    1. Wagner RC, Kreiner P, Barrnett RJ, Bitensky MW (1972) Biochemical characterization and cytochemical localization of a catecholamine-sensitive adenylate cyclase in isolated capillary endothelium. Proc Natl Acad Sci U S A 69(11):3175–3179

Source: PubMed

3
Subskrybuj