1
|
Onoki T, Kanczler J, Rawlings A, Smith M, Kim YH, Hashimoto K, Aizawa T, Oreffo ROC. Modulation of osteoblastogenesis by NRF2: NRF2 activation suppresses osteogenic differentiation and enhances mineralization in human bone marrow-derived mesenchymal stromal cells. FASEB J 2024; 38:e23892. [PMID: 39230563 DOI: 10.1096/fj.202400602r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
Mesenchymal stromal stem cells (MSCs) or skeletal stem cells (SSCs) play a major role in tissue repair due to their robust ability to differentiate into osteoblasts, chondrocytes, and adipocytes. Complex cell signaling cascades tightly regulate this differentiation. In osteogenic differentiation, Runt-related transcription factor 2 (RUNX2) and ALP activity are essential. Furthermore, during the latter stages of osteogenic differentiation, mineral formation mediated by the osteoblast occurs with the secretion of a collagenous extracellular matrix and calcium deposition. Activation of nuclear factor erythroid 2-related factor 2 (NRF2), an important transcription factor against oxidative stress, inhibits osteogenic differentiation and mineralization via modulation of RUNX2 function; however, the exact role of NRF2 in osteoblastogenesis remains unclear. Here, we demonstrate that NRF2 activation in human bone marrow-derived stromal cells (HBMSCs) suppressed osteogenic differentiation. NRF2 activation increased the expression of STRO-1 and KITLG (stem cell markers), indicating NRF2 protects HBMSCs stemness against osteogenic differentiation. In contrast, NRF2 activation enhanced mineralization, which is typically linked to osteogenic differentiation. We determined that these divergent results were due in part to the modulation of cellular calcium flux genes by NRF2 activation. The current findings demonstrate a dual role for NRF2 as a HBMSC maintenance factor as well as a central factor in mineralization, with implications therein for elucidation of bone formation and cellular Ca2+ kinetics, dystrophic calcification and, potentially, application in the modulation of bone formation.
Collapse
Affiliation(s)
- Takahiro Onoki
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Andrew Rawlings
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Melanie Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Ko Hashimoto
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Toshimi Aizawa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Mut J, Altmann S, Reising S, Meißner-Weigl J, Driessen MD, Ebert R, Seibel J. SiaNAl can be efficiently incorporated in glycoproteins of human mesenchymal stromal cells by metabolic glycoengineering. ACS Biomater Sci Eng 2024; 10:139-148. [PMID: 36946521 DOI: 10.1021/acsbiomaterials.2c01534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Metabolic glycoengineering involves the stimulation of cells with functionalized monosaccharides. Glucosamine, galactosamine, and mannosamine derivatives are commercially available, but their application may lead to undirected (i.e., chemical) incorporation into proteins. However, sialic acids are attached to the ends of complex sugar chains of glycoproteins, which might be beneficial for cell surface modification via click chemistry. Thus, we studied the incorporation of chemically synthesized unnatural alkyne modified sialic acid (SiaNAl) into glycoproteins of human telomerase-immortalized mesenchymal stromal cells (hMSC-TERT) and we show that SiaNAl can be efficiently incorporated in glycoproteins involved in signal transduction and cell junction.
Collapse
Affiliation(s)
- Jürgen Mut
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Stephan Altmann
- Department of Musculoskeletal Tissue Regeneration, University of Würzburg, Friedrich-Bergius-Ring 15, Würzburg 97076, Germany
| | - Sabine Reising
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Jutta Meißner-Weigl
- Department of Musculoskeletal Tissue Regeneration, University of Würzburg, Friedrich-Bergius-Ring 15, Würzburg 97076, Germany
| | - Marc D Driessen
- Institute for Molecular Medicine, Proteome Research, University Hospital and Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Regina Ebert
- Department of Musculoskeletal Tissue Regeneration, University of Würzburg, Friedrich-Bergius-Ring 15, Würzburg 97076, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| |
Collapse
|
3
|
Najar M, Melki R, Khalife F, Lagneaux L, Bouhtit F, Moussa Agha D, Fahmi H, Lewalle P, Fayyad-Kazan M, Merimi M. Therapeutic Mesenchymal Stem/Stromal Cells: Value, Challenges and Optimization. Front Cell Dev Biol 2022; 9:716853. [PMID: 35096805 PMCID: PMC8795900 DOI: 10.3389/fcell.2021.716853] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cellular therapy aims to replace damaged resident cells by restoring cellular and molecular environments suitable for tissue repair and regeneration. Among several candidates, mesenchymal stem/stromal cells (MSCs) represent a critical component of stromal niches known to be involved in tissue homeostasis. In vitro, MSCs appear as fibroblast-like plastic adherent cells regardless of the tissue source. The therapeutic value of MSCs is being explored in several conditions, including immunological, inflammatory and degenerative diseases, as well as cancer. An improved understanding of their origin and function would facilitate their clinical use. The stemness of MSCs is still debated and requires further study. Several terms have been used to designate MSCs, although consensual nomenclature has yet to be determined. The presence of distinct markers may facilitate the identification and isolation of specific subpopulations of MSCs. Regarding their therapeutic properties, the mechanisms underlying their immune and trophic effects imply the secretion of various mediators rather than direct cellular contact. These mediators can be packaged in extracellular vesicles, thus paving the way to exploit therapeutic cell-free products derived from MSCs. Of importance, the function of MSCs and their secretome are significantly sensitive to their environment. Several features, such as culture conditions, delivery method, therapeutic dose and the immunobiology of MSCs, may influence their clinical outcomes. In this review, we will summarize recent findings related to MSC properties. We will also discuss the main preclinical and clinical challenges that may influence the therapeutic value of MSCs and discuss some optimization strategies.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Rahma Melki
- Genetics and Immune-Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Ferial Khalife
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fatima Bouhtit
- Genetics and Immune-Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco.,Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Douaa Moussa Agha
- Genetics and Immune-Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco.,Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Mohammad Fayyad-Kazan
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Hadath, Lebanon.,Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Hadath, Lebanon
| | - Makram Merimi
- Genetics and Immune-Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco.,Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| |
Collapse
|
4
|
Luck J, Weil BD, Lowdell M, Mosahebi A. Adipose-Derived Stem Cells for Regenerative Wound Healing Applications: Understanding the Clinical and Regulatory Environment. Aesthet Surg J 2020; 40:784-799. [PMID: 31406975 DOI: 10.1093/asj/sjz214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is growing interest in the regenerative potential of adipose-derived stem cells (ADSCs) for wound healing applications. ADSCs have been shown to promote revascularization, activate local stem cell niches, reduce oxidative stress, and modulate immune responses. Combined with the fact that they can be harvested in large numbers with minimal donor site morbidity, ADSC products represent promising regenerative cell therapies. This article provides a detailed description of the defining characteristics and therapeutic potential of ADSCs, with a focus on understanding how ADSCs promote tissue regeneration and repair. It summarizes the current regulatory environment governing the use of ADSC products across Europe and the United States and examines how various adipose-derived products conform to the current UK legislative framework. Advice is given to clinicians and researchers on how novel ADSC therapeutics may be developed in accordance with regulatory guidelines.
Collapse
Affiliation(s)
| | - Benjamin D Weil
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital, London, UK
| | | | | |
Collapse
|
5
|
Klietz ML, Kückelhaus M, Kaiser HW, Raschke MJ, Hirsch T, Aitzetmüller M. Stammzellen in der Regenerativen Medizin – Translationale Hürden und Möglichkeiten zur Überwindung. HANDCHIR MIKROCHIR P 2020; 52:338-349. [DOI: 10.1055/a-1122-8916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ZusammenfassungDer Einsatz von mesenchymalen Stammzellen in der regenerativen Medizin wird immer populärer. Nichtsdestotrotz ist ihre Anwendung im klinischen Alltag noch immer limitiert. Zahlreiche ethische, rechtliche und translationale Probleme sowie Ungewissheit bzgl. der Sicherheit hemmen noch immer die Entstehung von entsprechenden Therapien aus vielversprechenden wissenschaftlichen Ansätzen.Diese Arbeit soll die Hauptprobleme bei der Translation von stammzellbasierten Therapien aus der Grundlagenforschung und Präklinik in den klinischen Alltag darstellen, sowie Ansätze aufzeigen, diese zu überwinden.
Collapse
Affiliation(s)
- Marie-Luise Klietz
- Abteilung für Plastische-, Rekonstruktive und Ästhetische Chirurgie, Handchirurgie, Fachklinik Hornheide, Münster
- Sektion Plastische Chirurgie an der Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Münster, Münster
- Abteilung für Plastische und Rekonstruktive Chirurgie, Institut für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster
| | - Maximilian Kückelhaus
- Abteilung für Plastische-, Rekonstruktive und Ästhetische Chirurgie, Handchirurgie, Fachklinik Hornheide, Münster
- Sektion Plastische Chirurgie an der Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Münster, Münster
- Abteilung für Plastische und Rekonstruktive Chirurgie, Institut für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster
| | | | - Michael J. Raschke
- Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Münster, Münster
| | - Tobias Hirsch
- Abteilung für Plastische-, Rekonstruktive und Ästhetische Chirurgie, Handchirurgie, Fachklinik Hornheide, Münster
- Sektion Plastische Chirurgie an der Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Münster, Münster
- Abteilung für Plastische und Rekonstruktive Chirurgie, Institut für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster
| | - Matthias Aitzetmüller
- Sektion Plastische Chirurgie an der Klinik für Unfall-, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Münster, Münster
- Abteilung für Plastische und Rekonstruktive Chirurgie, Institut für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster
| |
Collapse
|
6
|
Murine Mesenchymal Stromal Cells Retain Biased Differentiation Plasticity Towards Their Tissue of Origin. Cells 2020; 9:cells9030756. [PMID: 32204552 PMCID: PMC7140683 DOI: 10.3390/cells9030756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/15/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) reside in many human tissues and comprise a heterogeneous population of cells with self-renewal and multi-lineage differentiation potential, making them useful in regenerative medicine. It remains inconclusive whether MSCs isolated from different tissue sources exhibit variations in biological features. In this study, we derived MSCs from adipose tissue (AT-MSC) and compact bone (CB-MSC). We found that early passage of MSCs was readily expandable ex vivo, whereas the prolonged culture of MSCs showed alteration of cell morphology to fibroblastoid and reduced proliferation. CB-MSCs and AT-MSCs at passage 3 were CD29+, CD44+, CD105+, CD106+, and Sca-1+; however, passage 7 MSCs showed a reduction of MSC markers, indicating loss of stem cell population after prolonged culturing. Strikingly, CB-MSC was found more efficient at undergoing osteogenic differentiation, while AT-MSC was more efficient to differentiate into adipocytes. The biased differentiation pattern of MSCs from adipogenic or osteogenic tissue source was accompanied by preferential expression of the corresponding lineage marker genes. Interestingly, CB-MSCs treated with DNA demethylation agent 5-azacytidine showed enhanced osteogenic and adipogenic differentiation, whereas the treated AT-MSCs are less competent to differentiate. Our results suggest that the epigenetic state of MSCs is associated with the biased differentiation plasticity towards its tissue of origin, proposing a mechanism related to the retention of epigenetic memory. These findings facilitate the selection of optimal tissue sources of MSCs and the ex vivo expansion period for therapeutic applications.
Collapse
|
7
|
Use of Mesenchymal Stem/Stromal Cells for Pediatric Orthopedic Applications. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Brennen WN, Isaacs JT. Mesenchymal stem cells and the embryonic reawakening theory of BPH. Nat Rev Urol 2019; 15:703-715. [PMID: 30214054 DOI: 10.1038/s41585-018-0087-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The prostate is the only organ in a man that continues to grow with age. John McNeal proposed, 40 years ago, that this BPH is characterized by an age-related reinitiation of benign neoplastic growth selectively in developmentally abortive distal ducts within the prostate transition-periurethral zone (TPZ), owing to a reawakening of inductive stroma selectively within these zones. An innovative variant of this hypothesis is that, owing to its location, the TPZ is continuously exposed to urinary components and/or autoantigens, which produces an inflammatory TPZ microenvironment that promotes recruitment of bone marrow-derived mesenchymal stem cells (MSCs) and generates a paracrine-inductive stroma that reinitiates benign neoplastic nodular growth. In support of this hypothesis, MSCs infiltrate human BPH tissue and have the ability to stimulate epithelial stem cell growth. These results provide a framework for defining both the aetiology of BPH in ageing men and insights into new therapeutic approaches.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA. .,Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Bahsoun S, Coopman K, Forsyth NR, Akam EC. The Role of Dissolved Oxygen Levels on Human Mesenchymal Stem Cell Culture Success, Regulatory Compliance, and Therapeutic Potential. Stem Cells Dev 2018; 27:1303-1321. [DOI: 10.1089/scd.2017.0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
10
|
Sawicki LA, Choe LH, Wiley KL, Lee KH, Kloxin AM. Isolation and Identification of Proteins Secreted by Cells Cultured within Synthetic Hydrogel-Based Matrices. ACS Biomater Sci Eng 2018; 4:836-845. [PMID: 29552635 PMCID: PMC5850091 DOI: 10.1021/acsbiomaterials.7b00647] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/02/2018] [Indexed: 01/14/2023]
Abstract
Cells interact with and remodel their microenvironment, degrading large extracellular matrix (ECM) proteins (e.g., fibronectin, collagens) and secreting new ECM proteins and small soluble factors (e.g., growth factors, cytokines). Synthetic mimics of the ECM have been developed as controlled cell culture platforms for use in both fundamental and applied studies. However, how cells broadly remodel these initially well-defined matrices remains poorly understood and difficult to probe. In this work, we have established methods for widely examining both large and small proteins that are secreted by cells within synthetic matrices. Specifically, human mesenchymal stem cells (hMSCs), a model primary cell type, were cultured within well-defined poly(ethylene glycol) (PEG)-peptide hydrogels, and these cell-matrix constructs were decellularized and degraded for subsequent isolation and analysis of deposited proteins. Shotgun proteomics using liquid chromatography and mass spectrometry identified a variety of proteins, including the large ECM proteins fibronectin and collagen VI. Immunostaining and confocal imaging confirmed these results and provided visualization of protein organization within the synthetic matrices. Additionally, culture medium was collected from the encapsulated hMSCs, and a Luminex assay was performed to identify secreted soluble factors, including vascular endothelial growth factor (VEGF), endothelial growth factor (EGF), basic fibroblast growth factor (FGF-2), interleukin 8 (IL-8), and tumor necrosis factor alpha (TNF-α). Together, these methods provide a unique approach for studying dynamic reciprocity between cells and synthetic microenvironments and have the potential to provide new biological insights into cell responses during three-dimensional (3D) controlled cell culture.
Collapse
Affiliation(s)
- Lisa A. Sawicki
- Department
of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Leila H. Choe
- Department
of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
- Delaware
Biotechnology Institute, Newark, Delaware 19711, United States
| | - Katherine L. Wiley
- Department
of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Kelvin H. Lee
- Department
of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
- Delaware
Biotechnology Institute, Newark, Delaware 19711, United States
| | - April M. Kloxin
- Department
of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department
of Materials Science and Engineering, University
of Delaware, Newark, Delaware 19716, United
States
| |
Collapse
|
11
|
A High-Resolution Proteomic Landscaping of Primary Human Dental Stem Cells: Identification of SHED- and PDLSC-Specific Biomarkers. Int J Mol Sci 2018; 19:ijms19010158. [PMID: 29304003 PMCID: PMC5796107 DOI: 10.3390/ijms19010158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/25/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023] Open
Abstract
Dental stem cells (DSCs) have emerged as a promising tool for basic research and clinical practice. A variety of adult stem cell (ASC) populations can be isolated from different areas within the dental tissue, which, due to their cellular and molecular characteristics, could give rise to different outcomes when used in potential applications. In this study, we performed a high-throughput molecular comparison of two primary human adult dental stem cell (hADSC) sub-populations: Stem Cells from Human Exfoliated Deciduous Teeth (SHEDs) and Periodontal Ligament Stem Cells (PDLSCs). A detailed proteomic mapping of SHEDs and PDLSCs, via employment of nano-LC tandem-mass spectrometry (MS/MS) revealed 2032 identified proteins in SHEDs and 3235 in PDLSCs. In total, 1516 proteins were expressed in both populations, while 517 were unique for SHEDs and 1721 were exclusively expressed in PDLSCs. Further analysis of the recorded proteins suggested that SHEDs predominantly expressed molecules that are involved in organizing the cytoskeletal network, cellular migration and adhesion, whereas PDLSCs are highly energy-producing cells, vastly expressing proteins that are implicated in various aspects of cell metabolism and proliferation. Applying the Rho-GDI signaling pathway as a paradigm, we propose potential biomarkers for SHEDs and for PDLSCs, reflecting their unique features, properties and engaged molecular pathways.
Collapse
|
12
|
Peffers MJ, Collins J, Loughlin J, Proctor C, Clegg PD. A proteomic analysis of chondrogenic, osteogenic and tenogenic constructs from ageing mesenchymal stem cells. Stem Cell Res Ther 2016; 7:133. [PMID: 27624072 PMCID: PMC5022190 DOI: 10.1186/s13287-016-0384-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/28/2016] [Accepted: 08/05/2016] [Indexed: 02/02/2023] Open
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. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0384-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mandy J Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, CH64 7TE, UK. .,Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, The University of Liverpool, Leahurst, Neston, CH64 7TE, UK.
| | - John Collins
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, CH64 7TE, UK
| | - John Loughlin
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Carole Proctor
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, CH64 7TE, UK
| |
Collapse
|
13
|
Abstract
The past decade has seen an explosion of research directed toward better understanding of the mechanisms of mesenchymal stem/stromal cell (MSC) function during rescue and repair of injured organs and tissues. In addition to delineating cell–cell signaling and molecular controls for MSC differentiation, the field has made particular progress in defining several other mechanisms through which administered MSCs can promote tissue rescue/repair. These include: 1) paracrine activity that involves secretion of proteins/peptides and hormones; 2) transfer of mitochondria by way of tunneling nanotubes or microvesicles; and 3) transfer of exosomes or microvesicles containing RNA and other molecules. Improved understanding of MSC function holds great promise for the application of cell therapy and also for the development of powerful cell-derived therapeutics for regenerative medicine. Focusing on these three mechanisms, we discuss MSC-mediated effects on immune cell responses, cell survival, and fibrosis and review recent progress with MSC-based or MSC-derived therapeutics.
Collapse
Affiliation(s)
- Jeffrey L Spees
- University of Vermont, Burlington, VT, USA. .,Department of Medicine, Stem Cell Core, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT, 05446, USA.
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Texas A & M University College of Medicine, 206 Olsen Blvd., Room 228, MS1114, College Station, TX, 77845, USA
| | - Carl A Gregory
- Institute for Regenerative Medicine, Texas A & M University College of Medicine, 206 Olsen Blvd., Room 228, MS1114, College Station, TX, 77845, USA.
| |
Collapse
|
14
|
Segeletz S, Hoflack B. Proteomic approaches to study osteoclast biology. Proteomics 2016; 16:2545-2556. [PMID: 27350065 DOI: 10.1002/pmic.201500519] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/13/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022]
Abstract
Bone is a dynamic tissue whose remodeling throughout life is orchestrated by repeated cycles of destruction mediated by osteoclasts and rebuilding by osteoblasts. Current understanding of osteoclast biology has largely relied on the generation of knockout mice exhibiting an abnormal bone phenotype. This has provided a better understanding of osteoclast biology and the key proteins that support osteoclast function. However, mouse models alone do not provide an integrated view on protein networks and post-translational modifications that might be important for osteoclast function. During the past years, a number of MS-based quantitative methods have been developed to investigate the complexity of biological systems. This review will summarize how such approaches have contributed to the understanding of osteoclast differentiation and function.
Collapse
Affiliation(s)
- Sandra Segeletz
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Bernard Hoflack
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Gruber HE, Riley FE, Hoelscher GL, Ingram JA, Bullock L, Hanley EN. Human annulus progenitor cells: Analyses of this viable endogenous cell population. J Orthop Res 2016; 34:1351-60. [PMID: 27249627 DOI: 10.1002/jor.23319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/27/2016] [Indexed: 02/04/2023]
Abstract
Back pain and intervertebral disc degeneration have growing socioeconomic/health care impacts. Increasing research efforts address use of stem and progenitor cell-based replacement therapies to repopulate and regenerate the disc. Data presented here on the innate human annulus progenitor cells: (i) assessed osteogenic, chondrogenic and adipogenic potentials of cultured human annulus cells; and (ii) defined progenitor-cell related gene expression patterns. Verification of the presence of progenitor cells within primary human disc tissue also used immunohistochemical identification of cell surface markers and microarray analyses. Differentiation analysis in cell cultures demonstrated a viable progenitor cell pool within Thompson grades III-IV discs. Osteogenesis was present in 8 out of 11 cultures (73%), chondrogenesis in 8 of 11 (73%), and adipogenesis in 6 of 6 (100%). Immunolocalization was positive for CD29, CD44, CD105, and CD14 (mean values 80.2%, 81.5%, 85.1%, and 88.6%, respectively); localization of CD45 and CD34 was negative in disc tissue. Compared to controls, surgical discs showed significantly downregulated genes with recognized progenitor cell functions: TCF7L2 (2.7 fold), BMI1 (3.8 fold), FGF receptor 2 (2 fold), PAFAH1B1 (2.3 fold), and GSTP1 (9 fold). Compared to healthier grade I/II discs, grade III/IV discs showed significantly upregulated XRCC5 (3.6 fold), TCF7L2 (6 fold), GSTP1 (3.7 fold), and BMI1 (3 fold). Additional significant cell marker analyses showed expression of platelet-derived growth factor receptor alpha, CD90, CD73, and STRO-1. Statement of Clinical Significance: Findings provide the first identification of progenitor cells in annulus specimens from older, more degenerate discs (in contrast to earlier studies of healthier discs or nondegenerative specimens from teenagers). Findings also increase knowledge on progenitor cells present in the disc and suggest their value in potential future utilization for regeneration and disc cell therapy. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1351-1360, 2016.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Frank E Riley
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Gretchen L Hoelscher
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Jane A Ingram
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Letitia Bullock
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| | - Edward N Hanley
- Department of Orthopaedic Surgery, Carolinas Medical Center, Charlotte, North Calorina, 20226
| |
Collapse
|
16
|
Mukaihara K, Suehara Y, Kohsaka S, Kubota D, Toda-Ishii M, Akaike K, Fujimura T, Kobayashi E, Yao T, Ladanyi M, Kaneko K, Saito T. Expression of F-actin-capping protein subunit beta, CAPZB, is associated with cell growth and motility in epithelioid sarcoma. BMC Cancer 2016; 16:206. [PMID: 26965049 PMCID: PMC4787035 DOI: 10.1186/s12885-016-2235-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 03/01/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A previous proteomics study demonstrated the overexpression of F-actin capping protein subunit beta (CAPZB) in tissue specimens of epithelioid sarcoma (EpiS). The aim of the present study was to elucidate the function of CAPZB in EpiS. METHODS Cellular functional assays were performed in two EpiS cell lines using CAPZB siRNAs. In addition, comparative protein expression analyses using Isobaric Tags for Relative and Absolute Quantitation (i-TRAQ) method were performed to identify the specific proteins whose expression was dysregulated by CAPZB, and analysed the data with the Ingenuity Pathways Analysis (IPA) system using the obtained protein profiles to clarify the functional pathway networks associated with the oncogenic function of CAPZB in EpiS. Additionally, we performed functional assays of the INI1 protein using INI1-overexpressing EpiS cells. RESULTS All 15 EpiS cases showed an immunohistochemical expression of CAPZB, and two EpiS cell lines exhibited a strong CAPZB expression. Silencing of CAPZB inhibited the growth, invasion and migration of the EpiS cells. Analysis of protein profiles using the IPA system suggested that SWI/SNF chromatin-remodeling complexes including INI1 may function as a possible upstream regulator of CAPZB. Furthermore, silencing of CAPZB resulted in a decreased expression of INI1 proteins in the INI1-positive EpiS cells, whereas the induction of INI1 in the INI1-deficient EpiS cells resulted in an increased CAPZB mRNA expression. CONCLUSIONS CAPZB is involved in tumor progression in cases of EpiS, irrespective of the INI1 expression, and may be a potential therapeutic target. The paradoxical relationship between the tumor suppressor INI1 and the oncoprotein CAPZB in the pathogenesis of EpiS remains to be clarified.
Collapse
Affiliation(s)
- Kenta Mukaihara
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoshiyuki Suehara
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Shinji Kohsaka
- Department of Medical Genomics Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Daisuke Kubota
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Midori Toda-Ishii
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Human Pathology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Keisuke Akaike
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Human Pathology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tsutomu Fujimura
- Laboratory of Biochemical Analysis, Central Laboratory of Medical Sciences, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Eisuke Kobayashi
- Division of Musculoskeletal Oncology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takashi Yao
- Department of Human Pathology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Kazuo Kaneko
- Department of Orthopedic Surgery, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
17
|
Kasap M, Yeğenağa I, Akpinar G, Tuncay M, Aksoy A, Karaoz E. Comparative Proteome Analysis of hAT-MSCs Isolated from Chronic Renal Failure Patients with Differences in Their Bone Turnover Status. PLoS One 2015; 10:e0142934. [PMID: 26575497 PMCID: PMC4648497 DOI: 10.1371/journal.pone.0142934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/28/2015] [Indexed: 11/19/2022] Open
Abstract
The relationship between the stem cells and the bone turnover in uremic bone disease due to chronic renal failure (CRF) is not described. The aim of this study was to investigate the effect of bone turnover status on stem cell properties. To search for the presence of such link and shed some light on stem-cell relevant mechanisms of bone turnover, we carried out a study with mesenchymal stem cells. Tissue biopsies were taken from the abdominal subcutaneous adipose tissue of a CRF patient with secondary hyperparathyroidism with the high turnover bone disease. This patient underwent parathyroidectomy operation (PTX) and another sample was taken from this patient after PTX. A CRF patient with adynamic bone disease with low turnover and a healthy control were also included. Mesenchymal stem cells isolated from the subjects were analyzed using proteomic and molecular approaches. Except ALP activity, the bone turnover status did not affect common stem cell properties. However, detailed proteome analysis revealed the presence of regulated protein spots. A total of 32 protein spots were identified following 2D gel electrophoresis and MALDI-TOF/TOF analyzes. The identified proteins were classified into seven distinct groups and their potential relationship to bone turnover were discussed. Distinct protein expression patterns emerged in relation to the bone turnover status indicate a possible link between the stem cells and bone turnover in uremic bone disease due to CRF.
Collapse
MESH Headings
- Adipose Tissue/cytology
- Alkaline Phosphatase/metabolism
- Bone Diseases/complications
- Bone Diseases/diagnosis
- Bone Remodeling
- Cell Differentiation
- Cells, Cultured
- Electrophoresis, Gel, Two-Dimensional
- Humans
- Hyperparathyroidism, Secondary/complications
- Hyperparathyroidism, Secondary/diagnosis
- Hyperparathyroidism, Secondary/surgery
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/metabolism
- Parathyroidectomy
- Proteome/analysis
- Proteomics
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Telomerase/metabolism
Collapse
Affiliation(s)
- Murat Kasap
- Department of Medical Biology/DEKART Proteomics Laboratory, Kocaeli University Medical School, Kocaeli, Turkey
| | - Itır Yeğenağa
- Department of Internal Medicine, Nephrology, Kocaeli University Medical School, Kocaeli, Turkey
| | - Gurler Akpinar
- Department of Medical Biology/DEKART Proteomics Laboratory, Kocaeli University Medical School, Kocaeli, Turkey
| | - Mehmet Tuncay
- Department of Internal Medicine, Nephrology, Kocaeli University Medical School, Kocaeli, Turkey
| | - Ayça Aksoy
- Department of Stem Cell, Center for Stem Cell and Gene Therapies Research and Practice, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Erdal Karaoz
- Liv Hospital Center for Regenerative Medicine and Stem Cell Research and Manufacturing, İstanbul, Turkey
| |
Collapse
|
18
|
Song C, Wang J, Mo C, Mu S, Jiang X, Li X, Zhong S, Zhao Z, Zhou G. Use of Ferritin Expression, Regulated by Neural Cell-Specific Promoters in Human Adipose Tissue-Derived Mesenchymal Stem Cells, to Monitor Differentiation with Magnetic Resonance Imaging In Vitro. PLoS One 2015; 10:e0132480. [PMID: 26176961 PMCID: PMC4503445 DOI: 10.1371/journal.pone.0132480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/15/2015] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to establish a method for monitoring the neural differentiation of stem cells using ferritin transgene expression, under the control of a neural-differentiation-inducible promoter, and magnetic resonance imaging (MRI). Human adipose tissue-derived mesenchymal stem cells (hADMSCs) were transduced with a lentivirus containing the human ferritin heavy chain 1 (FTH1) gene coupled to one of three neural cell-specific promoters: human synapsin 1 promoter (SYN1p, for neurons), human glial fibrillary acidic protein promoter (GFAPp, for astrocytes), and human myelin basic protein promoter (MBPp, for oligodendrocytes). Three groups of neural-differentiation-inducible ferritin-expressing (NDIFE) hADMSCs were established: SYN1p-FTH1, GFAPp-FTH1, and MBPp-FTH1. The proliferation rate of the NDIFE hADMSCs was evaluated using a Cell Counting Kit-8 assay. Ferritin expression was assessed with western blotting and immunofluorescent staining before and after the induction of differentiation in NDIFE hADMSCs. The intracellular iron content was measured with Prussian blue iron staining and inductively coupled plasma mass spectrometry. R2 relaxation rates were measured with MRI in vitro. The proliferation rates of control and NDIFE hADMSCs did not differ significantly (P > 0.05). SYN1p-FTH1, GFAPp-FTH1, and MBPp-FTH1 hADMSCs expressed specific markers of neurons, astrocytes, and oligodendrocytes, respectively, after neural differentiation. Neural differentiation increased ferritin expression twofold, the intracellular iron content threefold, and the R2 relaxation rate two- to threefold in NDIFE hADMSCs, resulting in notable hypointensity in T2-weighted images (P < 0.05). These results were cross-validated. Thus, a link between neural differentiation and MRI signals (R2 relaxation rate) was established in hADMSCs. The use of MRI and neural-differentiation-inducible ferritin expression is a viable method for monitoring the neural differentiation of hADMSCs.
Collapse
Affiliation(s)
- Chengang Song
- Department of Anatomy, Institute of Clinical Anatomy, Southern Medical University, Guangzhou, China
| | - Jiachuan Wang
- School of Medicine, Shenzhen University, Shenzhen, China
- Department of Pathology, Shenzhen Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Cuiping Mo
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Shuhua Mu
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Xiaogang Jiang
- Department of Pathology, Southern Medical University, Guangzhou, China
| | - Xiaoyun Li
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, College of Life Sciences, South China Normal University, Guangzhou, China
| | - Shizhen Zhong
- Department of Anatomy, Institute of Clinical Anatomy, Southern Medical University, Guangzhou, China
- * E-mail: (SZ); (ZZ); (GZ)
| | - Zhenfu Zhao
- School of Medicine, Shenzhen University, Shenzhen, China
- * E-mail: (SZ); (ZZ); (GZ)
| | - Guangqian Zhou
- School of Medicine, Shenzhen University, Shenzhen, China
- * E-mail: (SZ); (ZZ); (GZ)
| |
Collapse
|
19
|
Somoza RA, Acevedo CA, Albornoz F, Luz-Crawford P, Carrión F, Young ME, Weinstein-Oppenheimer C. TGFβ3 secretion by three-dimensional cultures of human dental apical papilla mesenchymal stem cells. J Tissue Eng Regen Med 2015; 11:1045-1056. [PMID: 25690385 DOI: 10.1002/term.2004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 10/02/2014] [Accepted: 01/07/2015] [Indexed: 12/25/2022]
Abstract
Mesenchymal stem cells (MSCs) can be isolated from dental tissues, such as pulp and periodontal ligament; the dental apical papilla (DAP) is a less-studied MSC source. These dental-derived MSCs are of great interest because of their potential as an accessible source for cell-based therapies and tissue-engineering (TE) approaches. Much of the interest regarding MSCs relies on the trophic-mediated repair and regenerative effects observed when they are implanted. TGFβ3 is a key growth factor involved in tissue regeneration and scarless tissue repair. We hypothesized that human DAP-derived MSCs (hSCAPs) can produce and secrete TGFβ3 in response to micro-environmental cues. For this, we encapsulated hSCAPs in different types of matrix and evaluated TGFβ3 secretion. We found that dynamic changes of cell-matrix interactions and mechanical stress that cells sense during the transition from a monolayer culture (two-dimensional, 2D) towards a three-dimensional (3D) culture condition, rather than the different chemical composition of the scaffolds, may trigger the TGFβ3 secretion, while monolayer cultures showed almost 10-fold less secretion of TGFβ3. The study of these interactions is provided as a cornerstone in designing future strategies in TE and cell therapy that are more efficient and effective for repair/regeneration of damaged tissues. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rodrigo A Somoza
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Cristian A Acevedo
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Fernando Albornoz
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | | | - Flavio Carrión
- Laboratorio de Inmunología, Universidad de los Andes, Santiago, Chile
| | - Manuel E Young
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | | |
Collapse
|
20
|
Alegre-Aguarón E, Sampat SR, Xiong JC, Colligan RM, Bulinski JC, Cook JL, Ateshian GA, Brown LM, Hung CT. Growth factor priming differentially modulates components of the extracellular matrix proteome in chondrocytes and synovium-derived stem cells. PLoS One 2014; 9:e88053. [PMID: 24516581 PMCID: PMC3917883 DOI: 10.1371/journal.pone.0088053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 01/03/2014] [Indexed: 12/17/2022] Open
Abstract
To make progress in cartilage repair it is essential to optimize protocols for two-dimensional cell expansion. Chondrocytes and SDSCs are promising cell sources for cartilage repair. We previously observed that priming with a specific growth factor cocktail (1 ng/mL transforming growth factor-β1, 5 ng/mL basic fibroblast growth factor, and 10 ng/mL platelet-derived growth factor-BB) in two-dimensional culture, led to significant improvement in mechanical and biochemical properties of synovium-derived stem cell (SDSC)-seeded constructs. The current study assessed the effect of growth factor priming on the proteome of canine chondrocytes and SDSCs. In particular, growth factor priming modulated the proteins associated with the extracellular matrix in two-dimensional cultures of chondrocytes and SDSCs, inducing a partial dedifferentiation of chondrocytes (most proteins associated with cartilage were down-regulated in primed chondrocytes) and a partial differentiation of SDSCs (some collagen-related proteins were up-regulated in primed SDSCs). However, when chondrocytes and SDSCs were grown in pellet culture, growth factor-primed cells maintained their chondrogenic potential with respect to glycosaminoglycan and collagen production. In conclusion, the strength of the label-free proteomics technique is that it allows for the determination of changes in components of the extracellular matrix proteome in chondrocytes and SDSCs in response to growth factor priming, which could help in future tissue engineering strategies.
Collapse
Affiliation(s)
- Elena Alegre-Aguarón
- Department of Biomedical Engineering, Columbia University, New York, New York, United States of America
| | - Sonal R. Sampat
- Department of Biomedical Engineering, Columbia University, New York, New York, United States of America
| | - Jennifer C. Xiong
- Department of Biomedical Engineering, Columbia University, New York, New York, United States of America
| | - Ryan M. Colligan
- Quantitative Proteomics Center, Columbia University, New York, New York, United States of America
| | - J. Chloë Bulinski
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
| | - James L. Cook
- Comparative Orthopaedic Laboratory, University of Missouri, Columbia, Missouri, United States of America
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York, United States of America
- Department of Mechanical Engineering, Columbia University, New York, New York, United States of America
| | - Lewis M. Brown
- Quantitative Proteomics Center, Columbia University, New York, New York, United States of America
- * E-mail: (LMB); (CTH)
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York, United States of America
- * E-mail: (LMB); (CTH)
| |
Collapse
|
21
|
Zhu Z, Liu Z, Liu J, Bi M, Yang T, Wang J. Proteomic profiling of human placenta-derived mesenchymal stem cells upon transforming LIM mineralization protein-1 stimulation. Cytotechnology 2014; 67:285-97. [PMID: 24468833 DOI: 10.1007/s10616-013-9684-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 12/23/2013] [Indexed: 02/07/2023] Open
Abstract
Human placenta-derived mesenchymal stem cells (hPDMSCs) can differentiate into different types of cells and thus have tremendous potential for cell therapy and tissue engineering. LIM mineralization protein-1 (LMP-1) plays an important role in osteoblast differentiation, maturation and bone formation. To determine a global effect of LMP-1 on hPDMSCs, we designed a study using a proteomic approach combined with adenovirus-mediated gene transfer of LMP-1 to identify LMP-1-induced changes in hPDMSCs on proteome level. We have generated proteome maps of undifferentiated hPDMSCs and LMP-1 induced hPDMSCs. Two dimensional gel electrophoresis revealed 22 spots with at least 2.0-fold changes in expression and 15 differently expressed proteins were successfully identified by MALDI-TOF-MS. The proteins regulated by LMP-1 included cytoskeletal proteins, cadmium-binding proteins, and metabolic proteins, etc. The expression of some identified proteins was confirmed by further Western blot analyses. Our results will play an important role in better elucidating the underlying molecular mechanism in LMP-1 included hPDMSCs differentiation into osteoblasts.
Collapse
Affiliation(s)
- Zhen Zhu
- Stomatology Hospital, Jilin University, Changchun, 130021, People's Republic of China
| | | | | | | | | | | |
Collapse
|
22
|
Zimmerlin L, Park TS, Zambidis ET, Donnenberg VS, Donnenberg AD. Mesenchymal stem cell secretome and regenerative therapy after cancer. Biochimie 2013; 95:2235-45. [PMID: 23747841 PMCID: PMC3825748 DOI: 10.1016/j.biochi.2013.05.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/26/2013] [Indexed: 02/06/2023]
Abstract
Cancer treatment generally relies on tumor ablative techniques that can lead to major functional or disfiguring defects. These post-therapy impairments require the development of safe regenerative therapy strategies during cancer remission. Many current tissue repair approaches exploit paracrine (immunomodulatory, pro-angiogenic, anti-apoptotic and pro-survival effects) or restoring (functional or structural tissue repair) properties of mesenchymal stem/stromal cells (MSC). Yet, a major concern in the application of regenerative therapies during cancer remission remains the possible triggering of cancer recurrence. Tumor relapse implies the persistence of rare subsets of tumor-initiating cancer cells which can escape anti-cancer therapies and lie dormant in specific niches awaiting reactivation via unknown stimuli. Many of the components required for successful regenerative therapy (revascularization, immunosuppression, cellular homing, tissue growth promotion) are also critical for tumor progression and metastasis. While bi-directional crosstalk between tumorigenic cells (especially aggressive cancer cell lines) and MSC (including tumor stroma-resident populations) has been demonstrated in a variety of cancers, the effects of local or systemic MSC delivery for regenerative purposes on persisting cancer cells during remission remain controversial. Both pro- and anti-tumorigenic effects of MSC have been reported in the literature. Our own data using breast cancer clinical isolates have suggested that dormant-like tumor-initiating cells do not respond to MSC signals, unlike actively dividing cancer cells which benefited from the presence of supportive MSC. The secretome of MSC isolated from various tissues may partially diverge, but it includes a core of cytokines (i.e. CCL2, CCL5, IL-6, TGFβ, VEGF), which have been implicated in tumor growth and/or metastasis. This article reviews published models for studying interactions between MSC and cancer cells with a focus on the impact of MSC secretome on cancer cell activity, and discusses the implications for regenerative therapy after cancer.
Collapse
Affiliation(s)
- Ludovic Zimmerlin
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Tea Soon Park
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Elias T. Zambidis
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Vera S. Donnenberg
- University of Pittsburgh School of Medicine, Department of Cardiothoracic Surgery, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Albert D. Donnenberg
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Hematology/Oncology, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
23
|
Patil R, Kumar BM, Lee WJ, Jeon RH, Jang SJ, Lee YM, Park BW, Byun JH, Ahn CS, Kim JW, Rho GJ. Multilineage potential and proteomic profiling of human dental stem cells derived from a single donor. Exp Cell Res 2013; 320:92-107. [PMID: 24162002 DOI: 10.1016/j.yexcr.2013.10.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 10/03/2013] [Accepted: 10/05/2013] [Indexed: 02/08/2023]
Abstract
Dental tissues provide an alternative autologous source of mesenchymal stem cells (MSCs) for regenerative medicine. In this study, we isolated human dental MSCs of follicle, pulp and papilla tissue from a single donor tooth after impacted third molar extraction by excluding the individual differences. We then compared the morphology, proliferation rate, expression of MSC-specific and pluripotency markers, and in vitro differentiation ability into osteoblasts, adipocytes, chondrocytes and functional hepatocyte-like cells (HLCs). Finally, we analyzed the protein expression profiles of undifferentiated dental MSCs using 2DE coupled with MALDI-TOF-MS. Three types of dental MSCs largely shared similar morphology, proliferation potential, expression of surface markers and pluripotent transcription factors, and differentiation ability into osteoblasts, adipocytes, and chondrocytes. Upon hepatogenic induction, all MSCs were transdifferentiated into functional HLCs, and acquired hepatocyte functions by showing their ability for glycogen storage and urea production. Based on the proteome profiling results, we identified nineteen proteins either found commonly or differentially expressed among the three types of dental MSCs. In conclusion, three kinds of dental MSCs from a single donor tooth possessed largely similar cellular properties and multilineage potential. Further, these dental MSCs had similar proteomic profiles, suggesting their interchangeable applications for basic research and call therapy.
Collapse
Affiliation(s)
- Rajreddy Patil
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Brennen WN, Denmeade SR, Isaacs JT. Mesenchymal stem cells as a vector for the inflammatory prostate microenvironment. Endocr Relat Cancer 2013; 20:R269-90. [PMID: 23975882 PMCID: PMC3994592 DOI: 10.1530/erc-13-0151] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have an inherent tropism for sites of inflammation, which are frequently present in sites of cancer, including prostatic lesions. MSCs have been defined as CD73/CD90/CD105 triple-positive cells in the absence of hematopoietic lineage markers with the ability to differentiate into multiple mesodermal lineages, including osteoblasts, adipocytes, and chondrocytes. Our group has previously demonstrated that MSCs represent between 0.01 and 1.1% of the total cells present in human prostatectomy tissue. In addition to their multi-lineage differentiation potential, MSCs are immunoprivileged in nature and have a range of immunomodulatory effects on both the innate and adaptive arms of the immune system. MSCs have been detected in an increasing array of tissues, and evidence suggests that they are likely present in perivascular niches throughout the body. These observations suggest that MSCs represent critical mediators of the overall immune response during physiological homeostasis and likely contribute to pathophysiological conditions as well. Chronic inflammation has been suggested as an initiating event and progression factor in prostate carcinogenesis, a process in which the immunosuppressive properties of MSCs may play a role. MSCs have also been shown to influence malignant progression through a variety of other mechanisms, including effects on tumor proliferation, angiogenesis, survival, and metastasis. Additionally, human bone marrow-derived MSCs have been shown to traffic to human prostate cancer xenografts in immunocompromised murine hosts. The trafficking properties and immunoprivileged status of MSCs suggest that they can be exploited as an allogeneic cell-based vector to deliver cytotoxic or diagnostic agents for therapy.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
25
|
Proteome of human stem cells from periodontal ligament and dental pulp. PLoS One 2013; 8:e71101. [PMID: 23940696 PMCID: PMC3733711 DOI: 10.1371/journal.pone.0071101] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022] Open
Abstract
Background Many adult tissues contain a population of stem cells with the ability to regenerate structures similar to the microenvironments from which they are derived in vivo and represent a promising therapy for the regeneration of complex tissues in the clinical disorder. Human adult stem cells (SCs) including bone marrow stem cells (BMSCs), dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) have been characterized for their high proliferative potential, expression of characteristic SC-associated markers and for the plasticity to differentiate in different lineage in vitro. Methodology/Principal Findings The aim of this study is to define the molecular features of stem cells from oral tissue by comparing the proteomic profiles obtained with 2-DE followed by MALDI-TOF/TOF of ex-vivo cultured human PDLSCs, DPSCs and BMSCs. Our results showed qualitative similarities in the proteome profiles among the SCs examined including some significant quantitative differences. To enrich the knowledge of oral SCs proteome we performed an analysis in narrow range pH 4–7 and 6–9, and we found that DPSCs vs PDLSCs express differentially regulated proteins that are potentially related to growth, regulation and genesis of neuronal cells, suggesting that SCs derived from oral tissue source populations may possess the potential ability of neuronal differentiation which is very consistent with their neural crest origin. Conclusion/Significance This study identifies some differentially expressed proteins by using comparative analysis between DPSCs and PDLSCs and BMSCs and suggests that stem cells from oral tissue could have a different cell lineage potency compared to BMSCs.
Collapse
|
26
|
Salloum RH, Rubin JP, Marra KG. The role of steroids in mesenchymal stem cell differentiation: molecular and clinical perspectives. Horm Mol Biol Clin Investig 2013; 14:3-14. [DOI: 10.1515/hmbci-2013-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023]
Abstract
AbstractMesenchymal stem cells (MSCs) are multipotent stem cells capable of either self-regeneration or differentiation into more mature cell types, depending on the environmental stimuli. MSCs originate from the mesoderm and differentiate readily into mesodermal tissue. The tissues most studied in that respect are bone, fat and cartilage, and the key molecular elements in these three differentiation pathways are RUNX2, PPARγ and SOX9, respectively. Steroidal molecules play an important role in determining the fate of MSCs, mainly by altering the expression of key cellular molecules. Not all steroids exert the same effects on these cells. This review discusses the effects of sex steroids and glucocorticoids on the proliferative capacity and differentiation patterns of MSCs. With stem-cell-based therapy gaining worldwide attention, we explore the role of steroids in modulating MSCs for clinical and therapeutic purposes. The ease with which some MSCs, such as adipose-derived stem cells, can be harvested from the body and manipulated in the laboratory may lead to increased interest in this era of stem cells.
Collapse
|
27
|
Binato R, de Souza Fernandez T, Lazzarotto-Silva C, Du Rocher B, Mencalha A, Pizzatti L, Bouzas LF, Abdelhay E. Stability of human mesenchymal stem cells during in vitro culture: considerations for cell therapy. Cell Prolif 2012; 46:10-22. [PMID: 23163975 DOI: 10.1111/cpr.12002] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/08/2012] [Indexed: 01/30/2023] Open
Abstract
Ex vivo expansion and manipulation of human mesenchymal stem cells are important approaches to immunoregulatory and regenerative cell therapies. Although these cells show great potential for use, issues relating to their overall nature emerge as problems in the field. The need for extensive cell quantity amplification in vitro to obtain sufficient cell numbers for use, poses a risk of accumulating genetic and epigenetic abnormalities that could lead to sporadic malignant cell transformation. In this study, we have examined human mesenchymal stem cells derived from bone marrow, over extended culture time, using cytogenetic analyses, mixed lymphocyte reactions, proteomics and gene expression assays to determine whether the cultures would retain their potential for use in subsequent passages. Results indicate that in vitro cultures of these cells demonstrated chromosome variability after passage 4, but their immunomodulatory functions and differentiation capacity were maintained. At the molecular level, changes were observed from passage 5 on, indicating initiation of differentiation. Together, these results lead to the hypothesis that human mesenchymal stem cells cultures can be used successfully in cell therapy up to passage 4. However, use of cells from higher passages would have to be analysed case by case.
Collapse
Affiliation(s)
- R Binato
- Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Improved proteomic profiling of the cell surface of culture-expanded human bone marrow multipotent stromal cells. J Proteomics 2012; 78:1-14. [PMID: 23153793 DOI: 10.1016/j.jprot.2012.10.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/11/2012] [Accepted: 10/31/2012] [Indexed: 02/06/2023]
Abstract
A comprehensive analysis of the membrane proteome is essential to explain the biology of multipotent stromal cells and identify reliable protein biomarkers for the isolation as well as tracking of cells during differentiation and maturation. However, proteomic analysis of membrane proteins is challenging and they are noticeably under-represented in numerous proteomic studies. Here we introduce new approach, which includes high pressure-assisted membrane protein extraction, protein fractionation by gel-eluted liquid fraction entrapment electrophoresis (GELFREE), and combined use of liquid chromatography MALDI and ESI tandem mass spectrometry. This report presents the first comprehensive proteomic analysis of membrane proteome of undifferentiated and culture-expanded human bone marrow multipotent stromal cells (hBM-MSC) obtained from different human donors. Gene ontology mapping using the Ingenuity Pathway Analysis and DAVID programs revealed the largest membrane proteomic dataset for hBM-MSC reported to date. Collectively, the new workflow enabled us to identify at least two-fold more membrane proteins compared to published results on hBM-MSC. A total of 84 CDs were identified including 14 CDs identified for the first time. This dataset can serve as a basis for further exploration of self-renewal, differentiation and characterization of hBM-MSC.
Collapse
|
29
|
Rocha B, Calamia V, Mateos J, Fernández-Puente P, Blanco FJ, Ruiz-Romero C. Metabolic labeling of human bone marrow mesenchymal stem cells for the quantitative analysis of their chondrogenic differentiation. J Proteome Res 2012; 11:5350-61. [PMID: 22989065 DOI: 10.1021/pr300572r] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Human mesenchymal stem cells (hMSCs), residing in bone marrow as well as in the synovial lining of joints, can be triggered to differentiate toward chondrocytes. Thus, hMSCs harbor great therapeutic potential for the repair of cartilage defects in osteoarthritis (OA) and other articular diseases. However, the molecular mechanisms underlying the chondrogenesis process are still in part unknown. In this work, we applied for the first time the stable isotope labeling by amino acids in cell culture (SILAC) technique for the quantitative analysis of protein modulation during the chondrogenic differentiation process of hMSCs. First, we have standardized the metabolic labeling procedure on MSCs isolated from bone marrow (hBMSCs), and we have assessed the quality of chondrogenesis taking place in these conditions. Then, chondrogenic differentiation was induced on these labeled cells, and a quantitative proteomics approach has been followed to evaluate protein changes between two differentiation days. With this strategy, we could identify 622 different proteins by LC-MALDI-TOF/TOF analysis and find 65 proteins whose abundance was significantly modulated between day 2 and day 14 of chondrogenesis. Immunohistochemistry analyses were performed to verify the changes on a panel of six proteins that play different biological roles in the cell: fibronectin, gelsolin, vimentin, alpha-ATPase, mitochondrial superoxide dismutase, and cyclophilin A. All of these proteins were increased at day 14 compared to day 2 of chondrogenic induction, thus being markers of the enhanced extracellular matrix synthesis, cell adhesion, metabolism, and response to stress processes that take place in the early steps of chondrogenesis. Our strategy has allowed an additional insight into both specific protein function and the mechanisms of chondrogenesis and has provided a panel of protein markers of this differentiation process in hBMSCs.
Collapse
Affiliation(s)
- Beatriz Rocha
- Rheumatology Division, ProteoRed/ISCIII Proteomics Group, INIBIC-Hospital Universitario de A Coruña, 15006-A Coruña, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Critical role for lysyl oxidase in mesenchymal stem cell-driven breast cancer malignancy. Proc Natl Acad Sci U S A 2012; 109:17460-5. [PMID: 23033492 DOI: 10.1073/pnas.1206653109] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells with the ability to differentiate into multiple mesoderm lineages in the course of normal tissue homeostasis or during injury. We have previously shown that MSCs migrate to sites of tumorigenesis, where they become activated by cancer cells to promote metastasis. However, the molecular and phenotypic attributes of the MSC-induced metastatic state of the cancer cells remained undetermined. Here, we show that bone marrow-derived human MSCs promote de novo production of lysyl oxidase (LOX) from human breast carcinoma cells, which is sufficient to enhance the metastasis of otherwise weakly metastatic cancer cells to the lungs and bones. We also show that LOX is an essential component of the CD44-Twist signaling axis, in which extracellular hyaluronan causes nuclear translocation of CD44 in the cancer cells, thus triggering LOX transcription by associating with its promoter. Processed and enzymatically active LOX, in turn, stimulates Twist transcription, which mediates the MSC-triggered epithelial-to-mesenchymal transition (EMT) of carcinoma cells. Surprisingly, although induction of EMT in breast cancer cells has been tightly associated with the generation of cancer stem cells, we find that LOX, despite being critical for EMT, does not contribute to the ability of MSCs to promote the formation of cancer stem cells in the carcinoma cell populations. Collectively, our studies highlight a critical role for LOX in cancer metastasis and indicate that the signaling pathways controlling stroma-induced EMT are distinct from pathways regulating the development of cancer stem cells.
Collapse
|
31
|
Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev 2012; 21:2724-52. [PMID: 22468918 DOI: 10.1089/scd.2011.0722] [Citation(s) in RCA: 564] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) comprise a heterogeneous population of cells with multilineage differentiation potential, the ability to modulate oxidative stress, and secrete various cytokines and growth factors that can have immunomodulatory, angiogenic, anti-inflammatory and anti-apoptotic effects. Recent data indicate that these paracrine factors may play a key role in MSC-mediated effects in modulating various acute and chronic pathological conditions. MSCs are found in virtually all organs of the body. Bone marrow-derived MSCs (BM-MSCs) were discovered first, and the bone marrow was considered the main source of MSCs for clinical application. Subsequently, MSCs have been isolated from various other sources with the adipose tissue, serving as one of the alternatives to bone marrow. Adipose tissue-derived MSCs (ASCs) can be more easily isolated; this approach is safer, and also, considerably larger amounts of ASCs can be obtained compared with the bone marrow. ASCs and BM-MSCs share many biological characteristics; however, there are some differences in their immunophenotype, differentiation potential, transcriptome, proteome, and immunomodulatory activity. Some of these differences may represent specific features of BM-MSCs and ASCs, while others are suggestive of the inherent heterogeneity of both BM-MSC and ASC populations. Still other differences may simply be related to different isolation and culture protocols. Most importantly, despite the minor differences between these MSC populations, ASCs seem to be as effective as BM-MSCs in clinical application, and, in some cases, may be better suited than BM-MSCs. In this review, we will examine in detail the ontology, biology, preclinical, and clinical application of BM-MSCs versus ASCs.
Collapse
Affiliation(s)
- Marius Strioga
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | | | | | | | | |
Collapse
|
32
|
Peter S, Evans C, Ow SY, Scutt AM, Wright PC, Biggs CA. Proteomic analysis of the impact of static culturing on the expansion of rat bone marrow mesenchymal stem cells. Biotechnol Lett 2012; 34:1589-96. [DOI: 10.1007/s10529-012-0935-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 04/17/2012] [Indexed: 10/28/2022]
|
33
|
Tiwari A, Tursky ML, Mushahary D, Wasnik S, Collier FM, Suma K, Kirkland MA, Pande G. Ex vivo expansion of haematopoietic stem/progenitor cells from human umbilical cord blood on acellular scaffolds prepared from MS-5 stromal cell line. J Tissue Eng Regen Med 2012; 7:871-83. [PMID: 22511368 DOI: 10.1002/term.1479] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/18/2011] [Accepted: 01/16/2012] [Indexed: 12/13/2022]
Abstract
Lineage-specific expansion of haematopoietic stem/progenitor cells (HSPCs) from human umbilical cord blood (UCB) is desirable because of their several applications in translational medicine, e.g. treatment of cancer, bone marrow failure and immunodeficiencies. The current methods for HSPC expansion use either cellular feeder layers and/or soluble growth factors and selected matrix components coated on different surfaces. The use of cell-free extracellular matrices from bone marrow cells for this purpose has not previously been reported. We have prepared insoluble, cell-free matrices from a murine bone marrow stromal cell line (MS-5) grown under four different conditions, i.e. in presence or absence of osteogenic medium, each incubated under 5% and 20% O₂ tensions. These acellular matrices were used as biological scaffolds for the lineage-specific expansion of magnetically sorted CD34⁺ cells and the results were evaluated by flow cytometry and colony-forming assays. We could get up to 80-fold expansion of some HSPCs on one of the matrices and our results indicated that oxygen tension played a significant role in determining the expansion capacity of the matrices. A comparative proteomic analysis of the matrices indicated differential expression of proteins, such as aldehyde dehydrogenase and gelsolin, which have previously been identified as playing a role in HSPC maintenance and expansion. Our approach may be of value in identifying factors relevant to tissue engineering-based ex vivo HSPC expansion, and it may also provide insights into the constitution of the niche in which these cells reside in the bone marrow.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- CSIR Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India; Deakin University, Waurn Ponds, Geelong, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim HJ, Ji BR, Kim JS, Lee HN, Ha DH, Kim CW. Proteomic analysis of proteins associated with cellular senescence by calorie restriction in mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2012; 48:186-95. [DOI: 10.1007/s11626-012-9485-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/06/2012] [Indexed: 01/01/2023]
|
35
|
Williamson AJK, Whetton AD. The requirement for proteomics to unravel stem cell regulatory mechanisms. J Cell Physiol 2011; 226:2478-83. [PMID: 21792904 DOI: 10.1002/jcp.22610] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stem cells are defined by their ability to self-renew and to differentiate, the processes whereby these events are achieved is the subject of much investigation. These studies include cancer stem cell populations, where eradication of this specific population is the ultimate goal of treatment. Whilst cellular signalling events and transcription factor complex-mediated changes in gene expression have been analysed in some detail within stem cells, full systematic understanding of the events promoting self-renewal or the commitment process leading to formation of a specific cell type require a systems biology approach. This in turn demands a need for proteomic analysis of post-translational regulation of protein levels, protein interactions, protein post-translational modification (e.g. ubiquitination, methylation, acetylation, phosphorylation) to identify networks for stem cell regulation. Furthermore, the phenomenon of induced pluripotency via cellular reprogramming also can be understood optimally using combined molecular biology and proteomics approaches; here we describe current research employing proteomics and mass spectrometry to dissect stem cell regulatory mechanisms.
Collapse
Affiliation(s)
- Andrew J K Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, School of Cancer and Enabling Sciences, Manchester Academic Health Science Centre, The University of Manchester, Christie's NHS Foundation Trust, Wolfson Molecular Imaging Centre, Withington, Manchester, UK.
| | | |
Collapse
|
36
|
Rucevic M, Hixson D, Josic D. Mammalian plasma membrane proteins as potential biomarkers and drug targets. Electrophoresis 2011; 32:1549-64. [PMID: 21706493 DOI: 10.1002/elps.201100212] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining the plasma membrane proteome is crucial to understand the role of plasma membrane in fundamental biological processes. Change in membrane proteins is one of the first events that take place under pathological conditions, making plasma membrane proteins a likely source of potential disease biomarkers with prognostic or diagnostic potential. Membrane proteins are also potential targets for monoclonal antibodies and other drugs that block receptors or inhibit enzymes essential to the disease progress. Despite several advanced methods recently developed for the analysis of hydrophobic proteins and proteins with posttranslational modifications, integral membrane proteins are still under-represented in plasma membrane proteome. Recent advances in proteomic investigation of plasma membrane proteins, defining their roles as diagnostic and prognostic disease biomarkers and as target molecules in disease treatment, are presented.
Collapse
Affiliation(s)
- Marijana Rucevic
- COBRE Center for Cancer Research Development, Rhode Island Hospital, Providence, RI, USA
| | | | | |
Collapse
|
37
|
Maurer MH. Proteomic definitions of mesenchymal stem cells. Stem Cells Int 2011; 2011:704256. [PMID: 21437194 PMCID: PMC3062154 DOI: 10.4061/2011/704256] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 01/17/2011] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent cells isolated from the bone marrow and various other organs. They are able to proliferate and self-renew, as well as to give rise to progeny of at least the osteogenic, chondrogenic, and adipogenic lineages. Despite this functional definition, MSCs can also be defined by their expression of a distinct set of cell surface markers. In the current paper, studies investigating the proteome of human MSCs are reviewed with the aim to identify common protein markers of MSCs. The proteomic analysis of MSCs revealed a distinct set of proteins representing the basic molecular inventory, including proteins for (i) cell surface markers, (ii) the responsiveness to growth factors, (iii) the reuse of developmental signaling cascades in adult stem cells, (iv) the interaction with molecules of the extracellular matrix, (v) the expression of genes regulating transcription and translation, (vi) the control of the cell number, and (vii) the protection against cellular stress.
Collapse
Affiliation(s)
- Martin H Maurer
- Department of Physiology and Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
38
|
Mrozik KM, Zilm PS, Bagley CJ, Hack S, Hoffmann P, Gronthos S, Bartold PM. Proteomic characterization of mesenchymal stem cell-like populations derived from ovine periodontal ligament, dental pulp, and bone marrow: analysis of differentially expressed proteins. Stem Cells Dev 2011; 19:1485-99. [PMID: 20050811 DOI: 10.1089/scd.2009.0446] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Postnatal mesenchymal stem/stromal-like cells (MSCs) including periodontal ligament stem cells (PDLSCs), dental pulp stem cells (DPSCs), and bone marrow stromal cells (BMSCs) are capable of self-renewal and differentiation into multiple mesenchymal cell lineages. Despite their similar expression of MSC-associated and osteoblastic markers, MSCs retain the capacity to generate structures resembling the microenvironments from which they are derived in vivo and represent a promising therapy for the regeneration of complex tissues in the clinical setting. With this in mind, systematic approaches are required to identify the differential protein expression patterns responsible for lineage commitment and mediating the formation of these complex structures. This is the first study to compare the differential proteomic expression profiles of ex vivo-expanded ovine PDLSCs, DPSCs, and BMSCs derived from an individual donor. The two-dimensional electrophoresis was performed and regulated proteins were identified by liquid chromatography--electrospray-ionization tandem mass spectrometry (MS and MS/MS), database searching, and de novo sequencing. In total, 58 proteins were differentially expressed between at least 2 MSC populations in both sheep, 12 of which were up-regulated in one MSC population relative to the other two. In addition, the regulation of selected proteins was also conserved between equivalent human MSC populations. We anticipate that differential protein expression profiling will provide a basis for elucidating the protein expression patterns and molecular cues that are crucial in specifying the characteristic growth and developmental capacity of dental and non-dental tissue-derived MSC populations. These expression patterns can serve as important tools for the regeneration of particular tissues in future stem cell-based tissue engineering studies using animal models.
Collapse
Affiliation(s)
- Krzysztof M Mrozik
- Colgate Australian Clinical Dental Research Centre, Dental School, The University of Adelaide, Adelaide, Australia.
| | | | | | | | | | | | | |
Collapse
|
39
|
Wang W, Gou L, Yang J. What can proteomics teach us about bone marrow aging? Expert Rev Proteomics 2010; 7:799-802. [PMID: 21142879 DOI: 10.1586/epr.10.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Pivoriuūnas A, Surovas A, Borutinskaite V, Matuzeviccius D, Treigyte G, Savickiene J, Tunaitis V, Aldonyte R, Jarmalavicciuūte A, Suriakaite K, Liutkeviccius E, Venalis A, Navakauskas D, Navakauskiene R, Magnusson KE. Proteomic analysis of stromal cells derived from the dental pulp of human exfoliated deciduous teeth. Stem Cells Dev 2010; 19:1081-93. [PMID: 19824824 DOI: 10.1089/scd.2009.0315] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human dental pulp derived from exfoliated deciduous teeth has been described as a promising alternative source of multipotent stem cells. While these cells share certain similarities with mesenchymal stem-like cells (MSC) isolated from other tissues, basically they are still poorly characterized. In this study, for the first time, a proteomic map of abundantly expressed proteins in stromal cells derived from the dental pulp of human exfoliated deciduous teeth (SHED) was established. We also analyzed proteomic signatures of 2 clonal strains derived from SHEDs by single-cell cloning. The SHEDs were established from enzyme-disaggregated deciduous dental pulp from 6-year-old children. They had typical fibroblastoid morphology and high colony-forming efficiency index (16.4%). Cloning was performed at the second passage using limiting dilution in a 96-well plate (0.3 cell/well). Differentiation assessment revealed strong osteogenic but no adipogenic potential of the SHEDs in either clonal strain. The cells expressed characteristic antigens of MSC-like cells, including CD73, CD90, CD105, CD146, and did not express hematopoietic markers CD14, CD34, and CD45, as assessed with FACS analysis. For proteomic studies, cytosolic and nuclear proteins were analyzed with 2-dimensional gel electrophoresis (2-DE) and identified using matrix-assisted laser desorption/ionization (MALDI)-time of fl ight (TOF)-mass spectrometry (MS). All proteins were identified with high level of confidence (the lowest sequence coverage was 27%). Identification of highly expressed proteins in SHEDs revealed proteomic profiles very similar to that of MSC-like cells derived from other tissues. We also found a high degree of similarity between proteomic signatures of primary SHEDs and clonal cell strains. Thus, our data confirm a close resemblance between SHEDs and MSC-like cells from other tissues and may serve as starting point for creating-comprehensive proteomic maps.
Collapse
Affiliation(s)
- Augustas Pivoriuūnas
- Department of Experimental Medicine, Institute of Experimental and Clinical Medicine , Vilnius, Lithuania.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Moogk D, Stewart M, Gamble D, Bhatia M, Jervis E. Human ESC colony formation is dependent on interplay between self-renewing hESCs and unique precursors responsible for niche generation. Cytometry A 2010; 77:321-7. [PMID: 20217858 DOI: 10.1002/cyto.a.20878] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human embryonic stem cell (hESC) cultures are heterogeneous and constituting paracrine signals are required to maintain pluripotency. The cellular interplay and dynamic nature of this heterogeneity is not understood. Here, long-term hESC imaging and tracking revealed that hESC heterogeneity is dynamic and hESC self-renewal is dependent on colony-proximal distributions of paracrine signals. Tracking of hESCs forming colonies revealed that a biologically distinct cell type arises at the colony periphery in the absence of feeders. Higher rates of cell death occur in these hESC-derived cells, leading to clonal selection of colony reestablishing cells. hESC-derived feeders co-transferred during passaging promoted rapid colony recovery and expansion and reduced overall clonal selection of self-renewing hESCs. Our findings demonstrate that hESC-derived feeders arise from a distinct subpopulation of hESCs that respond to paracrine cues at the colony periphery that are required to sustain and establish clonal hESC self-renewal.
Collapse
Affiliation(s)
- Duane Moogk
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | | | | | | | |
Collapse
|
42
|
Abstract
Ectomesenchymal dental stem cells could be feasible tools for dental tissue engineering. Dental follicle cells are a promising example, since they are capable of differentiation into various dental tissue cells, such as osteoblasts or cementoblasts. However, cellular mechanisms of cell proliferation and differentiation are not understood in detail. Basic knowledge of these molecular processes may shorten the time before ectomesenchymal dental stem cells can be exploited for bone augmentation in regenerative medicine. Recent developments in proteomics and transcriptomics have made information about genome-wide expression profiles accessible, which can aid in clarifying molecular mechanisms of cells. This review describes the transcriptomes and proteomes of dental follicle cells before and after differentiation, and compares them with differentially expressed populations from dental tissue or bone marrow.
Collapse
Affiliation(s)
- C. Morsczeck
- Department of Operative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - G. Schmalz
- Department of Operative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
43
|
Ji YH, Ji JL, Sun FY, Zeng YY, He XH, Zhao JX, Yu Y, Yu SH, Wu W. 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; 9:550-64. [PMID: 20008835 PMCID: PMC2849707 DOI: 10.1074/mcp.m900243-mcp200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 12/14/2009] [Indexed: 11/06/2022] Open
Abstract
The chondrogenic potential of multipotent mesenchymal stem cells (MSCs) makes them a promising source for cell-based therapy of cartilage defects; however, the exact intracellular molecular mechanisms of chondrogenesis as well as self-renewal of MSCs remain largely unknown. To gain more insight into the underlying molecular mechanisms, we applied isobaric tag for relative and absolute quantitation (iTRAQ) labeling coupled with on-line two-dimensional LC/MS/MS technology to identify proteins differentially expressed in an in vitro model for chondrogenesis: chondrogenic differentiation of C3H10T1/2 cells, a murine embryonic mesenchymal cell line, was induced by micromass culture and 100 ng/ml bone morphogenetic protein 2 treatment for 6 days. A total of 1756 proteins were identified with an average false discovery rate <0.21%. Linear regression analysis of the quantitative data gave strong correlation coefficients: 0.948 and 0.923 for two replicate two-dimensional LC/MS/MS analyses and 0.881, 0.869, and 0.927 for three independent iTRAQ experiments, respectively (p < 0.0001). Among 1753 quantified proteins, 100 were significantly altered (95% confidence interval), and six of them were further validated by Western blotting. Functional categorization revealed that the 17 up-regulated proteins mainly comprised hallmarks of mature chondrocytes and enzymes participating in cartilage extracellular matrix synthesis, whereas the 83 down-regulated were predominantly involved in energy metabolism, chromatin organization, transcription, mRNA processing, signaling transduction, and cytoskeleton; except for a number of well documented proteins, the majority of these altered proteins were novel for chondrogenesis. Finally, the biological roles of BTF3l4 and fibulin-5, two novel chondrogenesis-related proteins identified in the present study, were verified in the context of chondrogenic differentiation. These data will provide valuable clues for our better understanding of the underlying mechanisms that modulate these complex biological processes and assist in the application of MSCs in cell-based therapy for cartilage regeneration.
Collapse
Affiliation(s)
- Yu-hua Ji
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Ministry of Education for Genetic Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
D'Alessandro A, Liumbruno G, Grazzini G, Pupella S, Lombardini L, Zolla L. Umbilical cord blood stem cells: Towards a proteomic approach. J Proteomics 2010; 73:468-82. [DOI: 10.1016/j.jprot.2009.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/04/2009] [Accepted: 06/16/2009] [Indexed: 02/07/2023]
|
45
|
Kuhn NZ, Tuan RS. Regulation of stemness and stem cell niche of mesenchymal stem cells: implications in tumorigenesis and metastasis. J Cell Physiol 2009; 222:268-77. [PMID: 19847802 DOI: 10.1002/jcp.21940] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human mesenchymal stem cells (MSCs) derived from adult tissues have been considered a candidate cell type for cell-based tissue engineering and regenerative medicine. These multipotent cells have the ability to differentiate along several mesenchymal lineages and possibly along non-mesenchymal lineages. MSCs possess considerable immunosuppressive properties that can influence the surrounding tissue positively during regeneration, but perhaps negatively towards the pathogenesis of cancer and metastasis. The balance between the naïve stem state and differentiation is highly dependent on the stem cell niche. Identification of stem cell niche components has helped to elucidate the mechanisms of stem cell maintenance and differentiation. Ultimately, the fate of stem cells is dictated by their microenvironment. In this review, we describe the identification and characterization of bone marrow-derived MSCs, the properties of the bone marrow stem cell niche, and the possibility and likelihood of MSC involvement in cancer progression and metastasis.
Collapse
Affiliation(s)
- Nastaran Z Kuhn
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | | |
Collapse
|
46
|
Lazzarotto-Silva C, Binato R, Rocher BD, Costa JACE, Pizzatti L, Bouzas LF, Abdelhay E. Similar proteomic profiles of human mesenchymal stromal cells from different donors. Cytotherapy 2009; 11:268-77. [PMID: 19333800 DOI: 10.1080/14653240902783268] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND AIMS Bone marrow (BM) stromal cells, also referred to as mesenchymal stromal cells (MSC), can be expanded ex vivo and are able to differentiate along multiple lineages, including chondrocytes, osteoblasts and adipocytes. MSC are known to secrete a number of cytokines and regulatory molecules implicated in different aspects of hematopoiesis, and seem to modulate the immune system. MSC appear to be promising candidates for cellular therapy associated with BM transplantation (BMT). METHODS We compared protein expression profiles of MSC cultures derived from different BM donors using two-dimensional (2-D) gel electrophoresis and matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) tandem mass spectrometry (MS/MS), and compared mixed lymphocyte reaction (MLR) assays in the absence and presence of third-party human (h) MSC derived from different donors during the same culture passage. RESULTS In a window of observation (pH 4-7, molecular weight 10-220 kDa), about 172 protein spots were obtained in each 2-D gel, corresponding to 84 distinct proteins. A comparative analysis demonstrated a very similar proteomic profile of cells of the first passage derived from different donors, suggesting that these cells have the same expression pattern. Additionally, cells derived from different donors were equally able to inhibit lymphocyte proliferation. CONCLUSIONS These results encourage the use of third-party MSC in cellular therapies, as cells derived from different individuals seem to have the same proteomic pattern and exhibit functionally similar properties.
Collapse
|
47
|
Kim BG, Lee JH, Ahn JM, Park SK, Cho JH, Hwang D, Yoo JS, Yates JR, Ryoo HM, Cho JY. ‘Two-Stage Double-Technique Hybrid (TSDTH)’ Identification Strategy for the Analysis of BMP2-Induced Transdifferentiation of Premyoblast C2C12 Cells to Osteoblast. J Proteome Res 2009; 8:4441-54. [DOI: 10.1021/pr900231a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Byung-Gyu Kim
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Ji-Hyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Jung-Mo Ahn
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Sung Kyu Park
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Ji-Hoon Cho
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Daehee Hwang
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Jong-Shin Yoo
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - John R. Yates
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Hyun-Mo Ryoo
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| | - Je-Yoel Cho
- Department of Biochemistry, School of Dentistry, Kyungpook National University, and 2nd BK21 program 700-422, Korea, Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92014, School of Interdisciplinary Bioscience and Bioengineering, Department of Chemical Engineering, POSTECH, Pohang, 790-784, Korea, Mass Spectrometer Development team, Korea Basic Science Institute, Daejeon, Korea, and Department of Cell and Developmental Biology, School of Dentistry, Seoul National
| |
Collapse
|
48
|
Copland IB, Lord-Dufour S, Cuerquis J, Coutu DL, Annabi B, Wang E, Galipeau J. Improved autograft survival of mesenchymal stromal cells by plasminogen activator inhibitor 1 inhibition. Stem Cells 2009; 27:467-77. [PMID: 19338064 DOI: 10.1634/stemcells.2008-0520] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) display robust reparative properties through their ability to limit apoptosis, enhance angiogenesis, and direct positive tissue remodeling. However, low in vivo survival of transplanted cells limits their overall effectiveness and significantly affects their clinical usage. Consequently, identifying strategies to improve cell survival in vivo are a priority. One explanation for their low survival is that MSCs are often transplanted into ischemic tissue, such as infarcted myocardium, where there is poor blood supply and low oxygen tension. Therefore, we examined how MSCs respond to a hypoxic, nutrient-poor stress environment to identify trophic factors that could be manipulated in advance of MSC transplantation. Combining microarray and proteomic screens we identified plasminogen activator inhibitor 1 (PAI-1) as one factor consistently upregulated in our in vitro ischemia-mimicking conditions. Subsequent genetic and chemical manipulation studies define PAI-1 as a negative regulator of MSC survival in vivo. Mechanistically, MSC-derived PAI-1 does not alter MSC survival through a plasmin-dependent mechanism but rather directly impacts on the adhesiveness of MSCs to their surrounding matrices. Thus we can conclude that post-transplantation, PAI-1 negatively impacts MSC survival by promoting anoikis via matrix detachment.
Collapse
Affiliation(s)
- Ian B Copland
- Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Mareddy S, Broadbent J, Crawford R, Xiao Y. Proteomic profiling of distinct clonal populations of bone marrow mesenchymal stem cells. J Cell Biochem 2009; 106:776-86. [PMID: 19229859 DOI: 10.1002/jcb.22088] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) have attracted immense research interest in the field of regenerative medicine due to their ability to be cultured for successive passages and multi-lineage differentiation. The molecular mechanisms governing MSC self-renewal and differentiation remain largely unknown. The development of sophisticated techniques, in particular clinical proteomics, has enabled researchers in various fields to identify and characterize cell specific biomarkers for therapeutic purposes. This study seeks to understand the cellular and sub-cellular processes responsible for the existence of stem cell populations in bone marrow samples by revealing the whole cell proteome of the clonal cultures of bone marrow-derived MSCs (BMSCs). Protein profiling of the MSC clonal populations was conducted by Two-Dimensional Liquid Chromatography/Matrix-Assisted Laser Desorption/Ionisation (MALDI) Mass Spectrometry (MS). A total of 83 proteins were identified with high confidence of which 11 showed differential expression between subpopulations, which included cytoskeletal and structural proteins, calcium binding proteins, cytokinetic proteins, and members of the intermediate filament family. This study generated a proteome reference map of BMSCs from the clonal populations, which will be valuable to better understand the underlying mechanism of BMSC self-renewal and differentiation.
Collapse
Affiliation(s)
- Shobha Mareddy
- Medical Device Domain, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, Brisbane, QLD 4059, Australia
| | | | | | | |
Collapse
|
50
|
Emerging molecular approaches in stem cell biology. Biotechniques 2009; 46:367-71. [PMID: 19480634 DOI: 10.2144/000113144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Stem cells are characterized by their ability to self-renew and differentiate into multiple adult cell types. Although substantial progress has been made over the last decade in understanding stem cell biology, recent technological advances in molecular and systems biology may hold the key to unraveling the mystery behind stem cell self-renewal and plasticity. The most notable of these advances is the ability to generate induced pluripotent cells from somatic cells. In this review, we discuss our current understanding of molecular similarities and differences among various stem cell types. Moreover, we survey the current state of systems biology and forecast future needs and direction in the stem cell field.
Collapse
|