A proteomic analysis of chondrogenic, osteogenic and tenogenic constructs from ageing mesenchymal stem cells

Mandy J Peffers, John Collins, John Loughlin, Carole Proctor, Peter D Clegg, Mandy J Peffers, John Collins, John Loughlin, Carole Proctor, Peter D Clegg

Abstract

Background: Mesenchymal stem cells (MSCs) have prospective applications in regenerative medicine and tissue engineering but to what extent phenotype and differentiation capacity alter with ageing is uncertain. Consequently, any loss in functionality with age would have profound consequences for the maintenance of tissue viability and the quality of tissues. Proteomics enables the set of proteins responsible for a particular cell phenotype to be identified, as well as enabling insights into mechanisms responsible for age-related alterations in musculoskeletal tissues. Few proteomic studies have been undertaken regarding age-related effects on tissue engineered into cartilage and bone, and none for tendon. This study provides a proteome inventory for chondrogenic, osteogenic and tenogenic constructs synthesised from human MSCs, and elucidates proteomic alterations as a consequence of donor age.

Methods: Human bone-marrow derived MSCs from young (n = 4, 21.8 years ± 2.4SD) and old (n = 4, 65.5 years ± 8.3SD) donors were used to make chondrogenic, osteogenic and tenogenic tissue-engineered constructs. We utilised an analytical method relying on extracted peptide intensities as a label-free approach for peptide quantitation by liquid chromatography-mass spectrometry. Results were validated using western blotting.

Results: We identified proteins that were differentially expressed with ageing; 128 proteins in chondrogenic constructs, 207 in tenogenic constructs and four in osteogenic constructs. Differentially regulated proteins were subjected to bioinformatic analysis to ascertain their molecular functions and the signalling pathways. For all construct types, age-affected proteins were involved in altered cell survival and death, and antioxidant and cytoskeletal changes. Energy and protein metabolism were the principle pathways affected in tenogenic constructs, whereas lipid metabolism was strongly affected in chondrogenic constructs and mitochondrial dysfunction in osteogenic constructs.

Conclusions: Our results imply that further work on MSC-based therapeutics for the older population needs to focus on oxidative stress protection. The differentially regulated proteome characterised by this study can potentially guide translational research specifically aimed at effective clinical interventions.

Keywords: Ageing; Chondrogenesis; Label-free quantification; Mesenchymal stem cells; Osteogenesis; Oxidative stress; Tenogenesis.

Figures

Fig. 1
Fig. 1
Histochemical and gene expression analysis of chondrogenic, osteogenic and tenogenic lineage differentiation for MSCs. Images are representative of all experiments. a MSC pellets cultured in control or chondrogenic media were fixed and stained with Alcian Blue (scale bar = 100 μm). Gene expression of b aggrecan, cCOL2A1 and dSOX9 following chondrogenic differentiation. Statistical evaluation was undertaken using Mann–Whitney U test (n = 6). e Osteogenic differentiation from MSCs was confirmed with Alizarin Red S staining at day 21 to visualise mineralised bone matrix following extraction of the calcified mineral from the stained monolayer at low pH. f Box and whisker plot showing quantitative results of Alizarin red staining. Statistical significance, Mann–Whitney U test p < 0.001 (n = 12). g Gene expression of RUNX2 following osteogenesis. h Histology images of a tendon construct made from MSCs stained with Masson’s Trichrome to identify collagenous matrix. Image was captured at ×4 magnification and ×10 magnification (inset, upper image) (scale bar = 100 μm). Example of more organised areas of collagen is marked on the inset image (red). (Lower image) Ultrastructural analysis using scanning TEM. Presence of aligned extracellular collagen fibrils (A) and less organised collagen (B) are inset (red) (scale bar = 1 μm). Tenogenic differentiation was also evaluated using gene expression of iCOL1A1, jSERPINF1 and kTHBS4. For gene expression, data are represented as 2–ΔCT compared with GAPDH. Statistical evaluation was undertaken using Mann–Whitney U test (n = 8) with data represented as 2–ΔCT compared with GAPDH. MSC mesenchymal stem cell
Fig. 2
Fig. 2
Coomassie-stained one-dimensional SDS-PAGE of the guanidine-soluble protein extracts of chrondrogenic and osteogenic constructs and Rapigest™ extracts of tenogenic constructs compared with Rapigest™ extract of MSCs. Images are representative of all experiments. Equal protein loading by weight (30 μg per well) allowed a qualitative and grossly quantitative comparison of soluble protein extracts. Vertical black line indicates independent gels. Lines indicate digital splicing. MSC mesenchymal stem cell
Fig. 3
Fig. 3
Pie charts depicting protein classification of DE proteins in ageing constructs using PANTHER. Proteins were demonstrated as DE when quantified with ProgenesisQI™ with at least two unique peptides, a 2-fold change in expression and q < 0.05. First row, chondrogenic constructs; second row, tenogenic constructs. a Biological processes, b molecular functions and c cellular components. Because osteogenic constructs had only four DE proteins with the filters of a 2-fold change in expression. p < 0.05 and q < 0.05, PANTHER analysis was undertaken on DE proteins quantified with ProgenesisQI™ with at least two unique peptides, a 2-fold change in expression and p < 0.05 (third row)
Fig. 4
Fig. 4
IPA generated networks derived from the proteins with different abundance in the chondrogenic, tenogenic and osteogenic constructs derived from young and old MSCs. IPA identified that lipid metabolism signalling pathways were predominant in chondrogenic constructs (a). In tenogenic constructs, signalling pathways were enriched for glucose metabolic processes (b). In osteogenic constructs, the principle signalling pathway was mitochondrial dysfunction (c). One of the principle functions associated with the DE proteins in tenogenic constructs was also protein metabolism (d). Green nodes, greater protein abundance in young; red nodes, greater protein abundance in old; white nodes, proteins not differentially abundant between young and old. Intensity of colour is related to higher fold-change. Key to the main features in the networks is shown
Fig. 5
Fig. 5
Western blotting validations of mass spectrometry results. COMP, biglycan and SOD1 abundance were confirmed by western blotting. Representative western blots for tenogenic constructs (a COMP and c biglycan) and chondrogenic constructs (e SOD1). Abundance of each protein is expressed semi-quantitatively (b, d, f) relative to total protein content. Statistical differences were assessed with age in the respective construct and antibody analysis using Mann–Whitney tests. Significant differences represented with p ≤ 0.05

References

    1. Pittenger MF, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–7. doi: 10.1126/science.284.5411.143.
    1. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med (Maywood) 2001;226(6):507–20.
    1. Kassem M, Abdallah BM. Human bone-marrow-derived mesenchymal stem cells: biological characteristics and potential role in therapy of degenerative diseases. Cell Tissue Res. 2008;331(1):157–63. doi: 10.1007/s00441-007-0509-0.
    1. Sethe S, Scutt A, Stolzing A. Aging of mesenchymal stem cells. Ageing Res Rev. 2006;5(1):91–116. doi: 10.1016/j.arr.2005.10.001.
    1. Stolzing A, et al. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev. 2008;129(3):163–73. doi: 10.1016/j.mad.2007.12.002.
    1. Laschober GT, et al. Age-specific changes of mesenchymal stem cells are paralleled by upregulation of CD106 expression as a response to an inflammatory environment. Rejuvenation Res. 2011;14(2):119–31. doi: 10.1089/rej.2010.1077.
    1. Zaim M, et al. Donor age and long-term culture affect differentiation and proliferation of human bone marrow mesenchymal stem cells. Ann Hematol. 2012;91(8):1175–86. doi: 10.1007/s00277-012-1438-x.
    1. Leskela HV, et al. Osteoblast recruitment from stem cells does not decrease by age at late adulthood. Biochem Biophys Res Commun. 2003;311(4):1008–13. doi: 10.1016/j.bbrc.2003.10.095.
    1. Alt EU, et al. Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res. 2012;8(2):215–25. doi: 10.1016/j.scr.2011.11.002.
    1. Okolicsanyi RK, et al. Human mesenchymal stem cells retain multilineage differentiation capacity including neural marker expression after extended in vitro expansion. PLoS One. 2015;10(9):e0137255. doi: 10.1371/journal.pone.0137255.
    1. Orth P, et al. Current perspectives in stem cell research for knee cartilage repair. Stem Cells Cloning. 2014;7:1–17.
    1. Gomez-Barrena E, et al. Bone fracture healing: cell therapy in delayed unions and nonunions. Bone. 2015;70:93–101. doi: 10.1016/j.bone.2014.07.033.
    1. Mazzocca AD, et al. Bone marrow-derived mesenchymal stem cells obtained during arthroscopic rotator cuff repair surgery show potential for tendon cell differentiation after treatment with insulin. Arthroscopy. 2011;27(11):1459–71. doi: 10.1016/j.arthro.2011.06.029.
    1. Docheva D, et al. Biologics for tendon repair. Adv Drug Deliv Rev. 2015;84:222–39. doi: 10.1016/j.addr.2014.11.015.
    1. Park HW, Shin JS, Kim CW. Proteome of mesenchymal stem cells. Proteomics. 2007;7(16):2881–94. doi: 10.1002/pmic.200700089.
    1. Sun HJ, et al. A proteomic analysis during serial subculture and osteogenic differentiation of human mesenchymal stem cell. J Orthop Res. 2006;24(11):2059–71. doi: 10.1002/jor.20273.
    1. Kurpinski K, et al. Proteomic profiling of mesenchymal stem cell responses to mechanical strain and TGF-beta1. Cell Mol Bioeng. 2009;2(4):606–14. doi: 10.1007/s12195-009-0090-6.
    1. Ji YH, et al. Quantitative proteomics analysis of chondrogenic differentiation of C3H10T1/2 mesenchymal stem cells by iTRAQ labeling coupled with on-line two-dimensional LC/MS/MS. Mol Cell Proteomics. 2010;9(3):550–64. doi: 10.1074/mcp.M900243-MCP200.
    1. Ju AHJ, Ko SP, Hyung SP. Membrane proteomic analysis of human mesenchymal stromal cells during adipogenesis. Proteomics. 2007;7(22):4181–91. doi: 10.1002/pmic.200700502.
    1. Peffers MJ, et al. Proteomic analysis reveals age-related changes in tendon matrix composition, with age- and injury-specific matrix fragmentation. J Biol Chem. 2014;289(37):25867–78. doi: 10.1074/jbc.M114.566554.
    1. Peffers MJ, Beynon RJ, Clegg PD. Absolute quantification of selected proteins in the human osteoarthritic secretome. Int J Mol Sci. 2013;14(10):20658–81. doi: 10.3390/ijms141020658.
    1. Fabre B, et al. Label-free quantitative proteomics reveals the dynamics of proteasome complexes composition and stoichiometry in a wide range of human cell lines. J Proteome Res. 2014;13(6):3027–37. doi: 10.1021/pr500193k.
    1. Kapacee Z, et al. Synthesis of embryonic tendon-like tissue by human marrow stromal/mesenchymal stem cells requires a three-dimensional environment and transforming growth factor beta3. Matrix Biol. 2010;29(8):668–77. doi: 10.1016/j.matbio.2010.08.005.
    1. Peffers MJ, et al. Age-related changes in mesenchymal stem cells identified using a multi-omics approach. Eur Cell Mater. 2016;31:136–59.
    1. Paul H, Reginato AJ, Schumacher HR. Alizarin red S staining as a screening test to detect calcium compounds in synovial fluid. Arthritis Rheum. 1983;26(2):191–200. doi: 10.1002/art.1780260211.
    1. Bancroft JD, Gamble M. Theory and practice of histological techniques. Philadelphia: Elsevier Health Sciences; 2008.
    1. Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987;1987(162):156–9. doi: 10.1016/0003-2697(87)90021-2.
    1. Peffers MJ, et al. Transcriptome analysis of ageing in uninjured human Achilles tendon. Arthritis Res Ther. 2015;17:33. doi: 10.1186/s13075-015-0544-2.
    1. Little D, et al. Proteomic differences between male and female anterior cruciate ligament and patellar tendon. PLoS One. 2014;9(5):e96526. doi: 10.1371/journal.pone.0096526.
    1. Peffers MJ, Thornton DJ, Clegg PD. Characterization of neopeptides in equine articular cartilage degradation. J Orthop Res. 2015;34:106–120.
    1. Vizcaino JA, et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat Biotechnol. 2014;32(3):223–6. doi: 10.1038/nbt.2839.
    1. Mi H, Muruganujan A, Thomas PD. PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res. 2013;41(Database issue):D377–86. doi: 10.1093/nar/gks1118.
    1. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. doi: 10.1038/nprot.2008.211.
    1. URL3. Ingenuity systems. Ingenuity pathway analysis.
    1. Hynes RO, Naba A. Overview of the matrisome—an inventory of extracellular matrix constituents and functions. Cold Spring Harb Perspect Biol. 2012;4(1):a004903. doi: 10.1101/cshperspect.a004903.
    1. Chen JQ, Wakefield LM, Goldstein DJ. Capillary nano-immunoassays: advancing quantitative proteomics analysis, biomarker assessment, and molecular diagnostics. J Transl Med. 2015;13:182. doi: 10.1186/s12967-015-0537-6.
    1. Oldershaw RA, et al. Notch signaling through Jagged-1 is necessary to initiate chondrogenesis in human bone marrow stromal cells but must be switched off to complete chondrogenesis. Stem Cells. 2008;26(3):666–74. doi: 10.1634/stemcells.2007-0806.
    1. Mueller AM. A systems biology approach to musculoskeletal tissue engineering: transcriptomic and proteomic analysis of cartilage and tendon cells, in musculoskeletal biology. Liverpool: University of Liverpool; 2015. p. 634.
    1. Peffers MJ. Personal communication. San Jose, CA, USA: ProteinSimple; 2016.
    1. Jelinsky SA, et al. Tendon-selective genes identified from rat and human musculoskeletal tissues. J Orthop Res. 2010;28(3):289–97.
    1. Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proc Natl Acad Sci U S A. 2003;100(16):9440–5. doi: 10.1073/pnas.1530509100.
    1. von der Mark K, et al. Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature. 1977;267(5611):531–2. doi: 10.1038/267531a0.
    1. Bosnakovski D, et al. Chondrogenic differentiation of bovine bone marrow mesenchymal stem cells in pellet cultural system. Exp Hematol. 2004;32(5):502–9. doi: 10.1016/j.exphem.2004.02.009.
    1. Kuo CK, Tuan RS. Mechanoactive tenogenic differentiation of human mesenchymal stem cells. Tissue Eng Part A. 2008;14(10):1615–27. doi: 10.1089/ten.tea.2006.0415.
    1. Cordonnier T, et al. 3D environment on human mesenchymal stem cells differentiation for bone tissue engineering. J Mater Sci Mater Med. 2010;21(3):981–7. doi: 10.1007/s10856-009-3916-9.
    1. Park E, Patel AN. Changes in the expression pattern of mesenchymal and pluripotent markers in human adipose-derived stem cells. Cell Biol Int. 2010;34(10):979–84. doi: 10.1042/CBI20100124.
    1. Peffers M. Protein and transcriptomic sigantures of cartilage ageing and disease, in Musculoskeletal Biology. Liverpool: University of Liverpool; 2013. p. 338.
    1. Bernstein P, et al. Expression pattern differences between osteoarthritic chondrocytes and mesenchymal stem cells during chondrogenic differentiation. Osteoarthr Cartil. 2010;18(12):1596–607. doi: 10.1016/j.joca.2010.09.007.
    1. Tiku ML, Shah R, Allison GT. Evidence linking chondrocyte lipid peroxidation to cartilage matrix protein degradation. Possible role in cartilage aging and the pathogenesis of osteoarthritis. J Biol Chem. 2000;275(26):20069–76. doi: 10.1074/jbc.M907604199.
    1. Greene MA, Loeser RF. Aging-related inflammation in osteoarthritis. Osteoarthr Cartil. 2015;23(11):1966–71. doi: 10.1016/j.joca.2015.01.008.
    1. Kasper G, et al. Insights into mesenchymal stem cell aging: involvement of antioxidant defense and actin cytoskeleton. Stem Cells. 2009;27(6):1288–97. doi: 10.1002/stem.49.
    1. Webb DJ, Parsons JT, Horwitz AF. Adhesion assembly, disassembly and turnover in migrating cells—over and over and over again. Nat Cell Biol. 2002;4(4):E97–100. doi: 10.1038/ncb0402-e97.
    1. Jin EJ, et al. Akt signaling regulates actin organization via modulation of MMP-2 activity during chondrogenesis of chick wing limb bud mesenchymal cells. J Cell Biochem. 2007;102(1):252–61. doi: 10.1002/jcb.21430.
    1. Xu B, et al. RhoA/ROCK, cytoskeletal dynamics, and focal adhesion kinase are required for mechanical stretch-induced tenogenic differentiation of human mesenchymal stem cells. J Cell Physiol. 2012;227(6):2722–9. doi: 10.1002/jcp.23016.
    1. Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev. 2015;24(10):1150–63. doi: 10.1089/scd.2014.0484.

Source: PubMed

3
Tilaa