Proteomic analysis of human mesenchymal stromal cell secretomes: a systematic comparison of the angiogenic potential

Debora Kehl, Melanie Generali, Anna Mallone, Manfred Heller, Anne-Christine Uldry, Phil Cheng, Benjamin Gantenbein, Simon P Hoerstrup, Benedikt Weber, Debora Kehl, Melanie Generali, Anna Mallone, Manfred Heller, Anne-Christine Uldry, Phil Cheng, Benjamin Gantenbein, Simon P Hoerstrup, Benedikt Weber

Abstract

Human mesenchymal stromal cell (hMSC) secretomes have shown to influence the microenvironment upon injury, promoting cytoprotection, angiogenesis, and tissue repair. The angiogenic potential is of particular interest for the treatment of ischemic diseases. Interestingly, hMSC secretomes isolated from different tissue sources have shown dissimilarities with respect to their angiogenic profile. This study compares angiogenesis of hMSC secretomes from adipose tissue (hADSCs), bone marrow (hBMSCs), and umbilical cord Wharton's jelly (hWJSCs). hMSC secretomes were obtained under xenofree conditions and analyzed by liquid chromatography tandem mass spectrometry (LC/MS-MS). Biological processes related to angiogenesis were found to be enriched in the proteomic profile of hMSC secretomes. hWJSC secretomes revealed a more complete angiogenic network with higher concentrations of angiogenesis related proteins, followed by hBMSC secretomes. hADSC secretomes lacked central angiogenic proteins and expressed most detected proteins to a significantly lower level. In vivo all secretomes induced vascularization of subcutaneously implanted Matrigel plugs in mice. Differences in secretome composition were functionally analyzed with monocyte and endothelial cell (EC) in vitro co-culture experiments using vi-SNE based multidimensional flow cytometry data analysis. Functional responses between hBMSC and hWJSC secretomes were comparable, with significantly higher migration of CD14++ CD16- monocytes and enhanced macrophage differentiation compared with hADSC secretomes. Both secretomes also induced a more profound pro-angiogenic phenotype of ECs. These results suggest hWJSCs secretome as the most potent hMSC source for inflammation-mediated angiogenesis induction, while the potency of hADSC secretomes was lowest. This systematic analysis may have implication on the selection of hMSCs for future clinical studies.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Characterization of hMSC from adipose tissue, bone marrow and umbilical cord Wharton’s jelly. a Trilineage differentiation of hMSCs (n = 5 per hMSC tissue source) into adipogenic (left; Oil Red O staining), osteogenic (middle; Alzarin Red S) and chondrogenic (right; Alcian Blue PAS) lineages. Scale bar = 100 μm. b Proliferation capacities of hMSCs (n = 5 per hMSC tissue source) over 12 days, c showing the cumulative population doublings of each hMSC source. d All hMSCs (n = 5 per hMSC tissue source) positively expressed CD73, CD90, CD105, and only a minimal proportion of cells were positive for CD14, CD34 and CD45. Data are visualized by bi-dimensional t-SNE maps generated from multidimensional flow cytometry data. e Recorded events were subdivided into 5 clusters by PhenoGraph and FlowSom algorithms according to surface marker expression. The cellular distribution of 5 donors per cell source is f visualized with cumulative density plots and g quantified accordingly. Bar graphs present mean ± s.d. (*p < 0.05, **p < 0.01, ***p < 0.001; one-way ANOVA and Tukey multiple comparison)
Fig. 2
Fig. 2
LC/MS-MS analysis of hMSC CM. a Heat maps (red = expressed, blue = not expressed) and b Venn diagrams represent the total amount of identified proteins in hMSC CM (n = 5 per hMSC tissue source), where more exclusive proteins and higher protein intensities were found for hWJSC CM. Identified proteins found in all hMSC CM were characterized on their d cellular location and e function
Fig. 3
Fig. 3
Enriched biological processes in hMSC CM. LC/MS-MS data were analyzed on overrepresented biological processes in the extracellular protein fraction of hMSC CM (n = 5 per hMSC tissue source). The most relevant biological processes, their protein-protein interactions and corresponding coloring patterns are displayed
Fig. 4
Fig. 4
The pro-angiogenic proteome of hMSC CM. a Heat map generated based on protein intensities displays hMSC CM proteins involved in angiogenesis (n = 5 per hMSC tissue source; red = expressed, blue = not expressed). b Interactions of angiogenesis related proteins, where node size/color depends on the amount of protein interactions. Proteins not found in the according CM are displayed in red, demonstrating that central pro-angiogenic proteins are missing in c hADSC CM compared to d hBMSC CM and e hWJSC CM. f The protein composition of hMSC CM and their exclusive proteins demonstrate that hBMSC CM and hWJSC CM are closer related compared to hADSC CM; g however significantly higher protein intensities by pairwise student t-test were found for hWJSC CM
Fig. 5
Fig. 5
Immune cell response to hMSC CM. a hMSC CM (n = 5 per tissue source) revealed enhanced migration of monocytes (n = 3 healthy blood donors), with significantly higher migration induced by hBMSC CM and hWJSC CM in comparison to hADSC CM and 5 ng/ml MCP-1 control. b Migrated monocytes were characterized by CD14 and CD16 staining, c demonstrating that hMSC CM mainly induced migration of classical CD14++ CD16− monocytes, but not of intermediate and non-classical monocytes. d 7 myeloid cell populations were identified after co-incubation of monocytic Thp-1 cells and hMSC CM (n = 5 per tissue source) displayed by a bi-dimensional cumulative vi-SNE map. e The percentages of each cell population were monitored, including f classical monocytes, g macrophages, and h all (diff.) (plasmacytoid) dendritic cells. hBMSC CM and hWJSC CM led to a significantly higher monocyte to macrophage differentiation in comparison to hADSC CM and basal DMEM control. Bar graphs present mean ± s.d. (*p < 0.05, **p < 0.01, ***p < 0.001; one-way ANOVA and Tukey multiple comparison)
Fig. 6
Fig. 6
hMSC secretome is pro-angiogenic in vitro and in vivo. a In vivo Matrigel plug assay demonstrated significantly increased vascularization when using hMSC CM compared to basal medium only. Scale bar = 0.5 cm / H&E = 100 μm. bd Stimulation of HUVEC with hMSC CM (n = 5 per tissue source) induces expression of angiogenesis related markers, including CD31, CD105, CD202b, and CD144. b The density distribution of cells was dependent on the co-incubation medium. Clustering flow cytometry data defined 4 different clusters, c each with a distinct expression profile. d A significantly higher shift towards the pro-angiogenic cluster 1 was found when HUVEC were co-incubated with hBMSC CM and hWJSC CM. Bar graphs present mean ± s.d. (*p < 0.05, **p < 0.01, ***p < 0.001; one-way ANOVA and Tukey multiple comparison)

References

    1. Galipeau J, Sensebe L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell. Stem. Cell. 2018;22:824–833. doi: 10.1016/j.stem.2018.05.004.
    1. Baraniak PR, McDevitt TC. Stem cell paracrine actions and tissue regeneration. Regen. Med. 2010;5:121–143. doi: 10.2217/rme.09.74.
    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. Tao H, Han Z, Han ZC, Li Z. Proangiogenic features of mesenchymal stem cells and their therapeutic applications. Stem Cells Int. 2016;2016:1314709. doi: 10.1155/2016/1314709.
    1. Kwon HM, et al. Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis. Vasc. Pharmacol. 2014;63:19–28. doi: 10.1016/j.vph.2014.06.004.
    1. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem. Cells Dev. 2015;24:1635–1647. doi: 10.1089/scd.2014.0316.
    1. Hsiao ST, et al. Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue. Stem. Cells Dev. 2012;21:2189–2203. doi: 10.1089/scd.2011.0674.
    1. Hsieh JY, et al. Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PLoS ONE. 2013;8:e72604. doi: 10.1371/journal.pone.0072604.
    1. Gaetani Massimiliano, Chinnici Cinzia Maria, Carreca Anna Paola, Di Pasquale Claudia, Amico Giandomenico, Conaldi Pier Giulio. Unbiased and quantitative proteomics reveals highly increased angiogenesis induction by the secretome of mesenchymal stromal cells isolated from fetal rather than adult skin. Journal of Tissue Engineering and Regenerative Medicine. 2017;12(2):e949–e961. doi: 10.1002/term.2417.
    1. Mallone A, Stenger C, Von Eckardstein A, Hoerstrup SP, Weber B. Biofabricating atherosclerotic plaques: In vitro engineering of a three-dimensional human fibroatheroma model. Biomaterials. 2018;150:49–59. doi: 10.1016/j.biomaterials.2017.09.034.
    1. Shen CC, et al. The angiogenic related functions of bone marrow mesenchymal stem cells are promoted by CBDL rat serum via the Akt/Nrf2 pathway. Exp. Cell Res. 2016;344:86–94. doi: 10.1016/j.yexcr.2016.04.013.
    1. Wang Y, et al. The transplantation of Akt-overexpressing amniotic fluid-derived mesenchymal stem cells protects the heart against ischemia-reperfusion injury in rabbits. Mol. Med. Rep. 2016;14:234–242. doi: 10.3892/mmr.2016.5212.
    1. Gnecchi M, et al. Early beneficial effects of bone marrow-derived mesenchymal stem cells overexpressing Akt on cardiac metabolism after myocardial infarction. Stem Cells. 2009;27:971–979. doi: 10.1002/stem.12.
    1. Kang K, et al. Exosomes secreted from CXCR4 overexpressing mesenchymal stem cells promote cardioprotection via Akt signaling pathway following myocardial infarction. Stem Cells Int. 2015;2015:659890. doi: 10.1155/2015/659890.
    1. Seghezzi G, et al. Fibroblast growth factor-2 (FGF-2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: an autocrine mechanism contributing to angiogenesis. J. Cell. Biol. 1998;141:1659–1673. doi: 10.1083/jcb.141.7.1659.
    1. Lee TJ, et al. Enhancement of long-term angiogenic efficacy of adipose stem cells by delivery of FGF2. Microvasc. Res. 2012;84:1–8. doi: 10.1016/j.mvr.2012.04.004.
    1. Lopatina T, et al. The angiogenic potential of adipose mesenchymal stem cell-derived extracellular vesicles is modulated by basic fibroblast growth factor. J. Stem Cell Res. Ther. 2014;4:245.
    1. Pires AO, et al. Unveiling the differences of secretome of human bone marrow mesenchymal stem cells, adipose tissue-derived stem cells, and human umbilical cord perivascular cells: a proteomic analysis. Stem. Cells Dev. 2016;25:1073–1083. doi: 10.1089/scd.2016.0048.
    1. Hsieh JY, Fu YS, Chang SJ, Tsuang YH, Wang HW. Functional module analysis reveals differential osteogenic and stemness potentials in human mesenchymal stem cells from bone marrow and Wharton’s jelly of umbilical cord. Stem. Cells Dev. 2010;19:1895–1910. doi: 10.1089/scd.2009.0485.
    1. Corliss BA, Azimi MS, Munson JM, Peirce SM, Murfee WL. Macrophages: an inflammatory link between angiogenesis and lymphangiogenesis. Microcirculation. 2016;23:95–121. doi: 10.1111/micc.12259.
    1. Moldovan NI. Role of monocytes and macrophages in adult angiogenesis: a light at the tunnel’s end. J. Hematother. Stem Cell Res. 2002;11:179–194. doi: 10.1089/152581602753658394.
    1. Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006;8:315–317. doi: 10.1080/14653240600855905.
    1. Mendicino M, Bailey AM, Wonnacott K, Puri RK, Bauer SR. MSC-based product characterization for clinical trials: an FDA perspective. Cell. Stem. Cell. 2014;14:141–145. doi: 10.1016/j.stem.2014.01.013.
    1. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 2016;25:829–848. doi: 10.3727/096368915X689622.
    1. Brown KJ, et al. Advances in the proteomic investigation of the cell secretome. Expert. Rev. Proteom. 2012;9:337–345. doi: 10.1586/epr.12.21.
    1. Lee RH, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell. Stem. Cell. 2009;5:54–63. doi: 10.1016/j.stem.2009.05.003.
    1. Sun CK, et al. Mixed serum-deprived and normal adipose-derived mesenchymal stem cells against acute lung ischemia-reperfusion injury in rats. Am. J. Transl. Res. 2015;7:209–231.
    1. Ozcan S, et al. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging. 2016;8:1316–1329. doi: 10.18632/aging.100971.
    1. von Bahr L, et al. Long-term complications, immunologic effects, and role of passage for outcome in mesenchymal stromal cell therapy. Biol. Blood. Marrow Transplant. 2012;18:557–564. doi: 10.1016/j.bbmt.2011.07.023.
    1. di Fagagna FD. Living on a break: cellular senescence as a DNA-damage response. Nat. Rev. Cancer. 2008;8:512–522. doi: 10.1038/nrc2440.
    1. Dominguez-Sola D, et al. Non-transcriptional control of DNA replication by c-Myc. Nature. 2007;448:445–451. doi: 10.1038/nature05953.
    1. Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell. Stem. Cell. 2012;10:244–258. doi: 10.1016/j.stem.2012.02.005.
    1. Ranganath RM. Harnessing the developmental potential of nucellar cells: barriers and opportunities. Trends Biotechnol. 2004;22:504–510. doi: 10.1016/j.tibtech.2004.08.010.
    1. Zhou BR, et al. The effect of conditioned media of adipose-derived stem cells on wound healing after ablative fractional carbon dioxide laser resurfacing. Biomed. Res. Int. 2013;2013:519126.
    1. Kordelas L, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28:970–973. doi: 10.1038/leu.2014.41.
    1. Shin H, Ryu HH, Kwon O, Park BS, Jo SJ. Clinical use of conditioned media of adipose tissue-derived stem cells in female pattern hair loss: a retrospective case series study. Int. J. Dermatol. 2015;54:730–735. doi: 10.1111/ijd.12650.
    1. Fukuoka H, Suga H. Hair regeneration treatment using adipose-derived stem cell conditioned medium: follow-up with trichograms. Eplasty. 2015;15:e10.
    1. Dahbour S, et al. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci. Ther. 2017;23:866–874. doi: 10.1111/cns.12759.
    1. Katagiri W, Osugi M, Kawai T, Hibi H. First-in-human study and clinical case reports of the alveolar bone regeneration with the secretome from human mesenchymal stem cells. Head Face Med. 2016;12:5. doi: 10.1186/s13005-016-0101-5.
    1. Kehl D, et al. Amniotic fluid cells show higher pluripotency-related gene expression than allantoic fluid cells. Stem Cells Dev. 2017;26:1424–1437. doi: 10.1089/scd.2016.0352.
    1. Bieback K, et al. Human alternatives to fetal bovine serum for the expansion of mesenchymal stromal cells from bone marrow. Stem Cells. 2009;27:2331–2341. doi: 10.1002/stem.139.
    1. Amir el AD, et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 2013;31:545–552. doi: 10.1038/nbt.2594.
    1. Svatek RS, et al. A multiplexed, particle-based flow cytometric assay identified plasma matrix metalloproteinase-7 to be associated with cancer-related death among patients with bladder cancer. Cancer. 2010;116:4513–4519. doi: 10.1002/cncr.25401.
    1. Gazdhar A, et al. Alpha-Klotho enrichment in induced pluripotent stem cell secretome contributes to antioxidative protection in acute lung injury. Stem Cells. 2018;36:616–625. doi: 10.1002/stem.2752.
    1. Brugger V, et al. Delaying histone deacetylase response to injury accelerates conversion into repair Schwann cells and nerve regeneration. Nat. Commun. 2017;8:14272. doi: 10.1038/ncomms14272.
    1. Lazar C, Gatto L, Ferro M, Bruley C, Burger T. Accounting for the multiple natures of missing values in label-free quantitative proteomics data sets to compare imputation strategies. J. Proteome Res. 2016;15:1116–1125. doi: 10.1021/acs.jproteome.5b00981.

Source: PubMed

3
Subskrybuj