1
|
Vadhan A, Gupta T, Hsu WL. Mesenchymal Stem Cell-Derived Exosomes as a Treatment Option for Osteoarthritis. Int J Mol Sci 2024; 25:9149. [PMID: 39273098 PMCID: PMC11395657 DOI: 10.3390/ijms25179149] [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] [Received: 07/30/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Osteoarthritis (OA) is a leading cause of pain and disability worldwide in elderly people. There is a critical need to develop novel therapeutic strategies that can effectively manage pain and disability to improve the quality of life for older people. Mesenchymal stem cells (MSCs) have emerged as a promising cell-based therapy for age-related disorders due to their multilineage differentiation and strong paracrine effects. Notably, MSC-derived exosomes (MSC-Exos) have gained significant attention because they can recapitulate MSCs into therapeutic benefits without causing any associated risks compared with direct cell transplantation. These exosomes help in the transport of bioactive molecules such as proteins, lipids, and nucleic acids, which can influence various cellular processes related to tissue repair, regeneration, and immune regulation. In this review, we have provided an overview of MSC-Exos as a considerable treatment option for osteoarthritis. This review will go over the underlying mechanisms by which MSC-Exos may alleviate the pathological hallmarks of OA, such as cartilage degradation, synovial inflammation, and subchondral bone changes. Furthermore, we have summarized the current preclinical evidence and highlighted promising results from in vitro and in vivo studies, as well as progress in clinical trials using MSC-Exos to treat OA.
Collapse
Affiliation(s)
- Anupama Vadhan
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin 632007, Taiwan;
| | - Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan;
| | - Wen-Li Hsu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin 632007, Taiwan;
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
2
|
Walentowicz P, Sadlecki P, Walentowicz-Sadlecka M, Bajek A, Grabiec M, Drewa T. Human amniotic fluid as a source of stem cells. Open Med (Wars) 2022; 17:648-660. [PMID: 35434378 PMCID: PMC8982042 DOI: 10.1515/med-2022-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Human amniotic fluid collected during amniocentesis contains a heterogeneous population of differentiated and undifferentiated cells. Properties and number of these cells vary depending on the gestational age and the presence of potential fetal pathologies. The aim of this study was to analyze the effects of maternal, fetal, and environmental factors on the success rates of amniotic fluid stem cell cultures, the number of human amniotic fluid stem cells (hAFSC), their growth rates in primary cultures, and the number of cell passages. The study included 355 patients qualified for genetic amniocentesis at the Prenatal Genetic Unit, Department of Obstetrics, Gynecology and Oncologic Gynecology, Nicolaus Copernicus University Medical College in Bydgoszcz in 2011–2017. The mean age of the study participants was 34 ± 6.2 years, and mean gravidity amounted to 2.48 ± 1.4. Amniotic fluid sample volume turned out to be a highly significant (p < 0.01) predictor of culture success, and the relationship was particularly evident in women older than 40 years. Another highly significant predictor of culture success was the presence of two cell populations in the sample (p < 0.01). The likelihood of culture success correlated significantly (p < 0.05) with the season of the year at the time of amniocentesis. The number of cell passages differed significantly depending on the maternal age (p < 0.01). The number of passages also showed a highly significant relationship with the season of the year the sample was obtained (p < 0.01). Younger maternal age was identified as a determinant of high passage number (≥3), and another highly significant determinant of high passage number was the presence of two cell populations in the amniotic fluid sample (p < 0.01). Percentage of successfully established hAFSC cultures and the number of passages depended on amniotic fluid volume, the presence of two cell populations within the sample, and the season of the year. Individual characteristics of the donors, such as age and gravidity, did not exert a significant effect on the number of isolated hAFSCs and the rate of their growth. Patients’ place of residence, fetal karyotype, transportation time, and purity of the samples did not affect the success rates for primary cultures and the number of passages.
Collapse
Affiliation(s)
- Pawel Walentowicz
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- Department of Obstetrics, Gynecology and Oncological Gynecology, Regional Polyclinical Hospital , 87-100 Torun , Poland
| | - Pawel Sadlecki
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- Department of Obstetrics, Gynecology and Oncological Gynecology, Regional Polyclinical Hospital , 87-100 Torun , Poland
| | - Malgorzata Walentowicz-Sadlecka
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- 2nd Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education , 01-809 Warsaw , Poland
| | - Anna Bajek
- Department of Tissue Engineering, Nicolaus Copernicus University , Bydgoszcz 85-092 , Poland
| | - Marek Grabiec
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
| | - Tomasz Drewa
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz , 85-094 , Poland
| |
Collapse
|
3
|
Barisic D, Erb M, Follo M, Al-Mudaris D, Rolauffs B, Hart ML. Lack of a skeletal muscle phenotype in adult human bone marrow stromal cells following xenogeneic-free expansion. Stem Cell Res Ther 2020; 11:79. [PMID: 32087752 PMCID: PMC7036219 DOI: 10.1186/s13287-020-1587-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/22/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Many studies have elegantly shown that murine and rat bone marrow-derived mesenchymal stromal cells (bmMSCs) contribute to muscle regeneration and improve muscle function. Yet, the ability of transplanted human bmMSCs to manifest myogenic potential shows conflicting results. While human adipose- and umbilical cord-derived MSCs can be differentiated into a skeletal muscle phenotype using horse serum (HS), bmMSCs have only been shown to differentiate towards the skeletal muscle lineage using a complex mixture of cytokines followed by transfection with notch intracellular domain. Methods Since xenogeneic-free growth supplements are increasingly being used in the expansion of bmMSCs in clinical trials, we investigated the effects of human plasma and platelet lysate (P/PL) on the expression of neuromuscular markers and whether P/PL-expanded human bmMSCs could be differentiated towards a skeletal myogenic phenotype. Neuromuscular markers were measured using the highly sensitive droplet digital polymerase chain reaction for measuring the expression of Myf5, MyoD, MyoG, ACTA1, Desmin, GAP-43, and Coronin 1b transcripts, by performing immunofluorescence for the expression of Desmin, GAP-43, and MEF2, and flow cytometry for the expression of CD56/neural cell adhesion molecule (NCAM). Results Despite that bmMSCs expressed the myogenic regulatory factor (MRF) MEF2 after expansion in P/PL, bmMSCs cultured under such conditions did not express other essential MRFs including Myf5, MyoD, MyoG, or ACTA1 needed for myogenesis. Moreover, HS did not induce myogenesis of bmMSCs and hence did not induce the expression of any of these myogenic markers. P/PL, however, did lead to a significant increase in neurogenic GAP-43, as well as Desmin expression, and resulted in a high baseline expression of the neurogenic gene Coronin 1b which was sustained under further P/PL or HS culture conditions. Fetal bovine serum resulted in equally high levels of GAP-43 and Coronin 1b. Moreover, the proportion of CD56/NCAM-positive bmMSCs cultured in P/PL was 5.9 ± 2.1. Conclusions These data suggest that P/PL may prime a small portion of bmMSCs towards an early neural precursor cell type. Collectively, this shows that P/PL partially primes the cells towards a neurogenic phenotype, but does not prime adult human bmMSCs towards the skeletal muscle lineage.
Collapse
Affiliation(s)
- Dominik Barisic
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marita Erb
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dahlia Al-Mudaris
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie L Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Vasilev G, Ivanova M, Ivanova-Todorova E, Tumangelova-Yuzeir K, Krasimirova E, Stoilov R, Kyurkchiev D. Secretory factors produced by adipose mesenchymal stem cells downregulate Th17 and increase Treg cells in peripheral blood mononuclear cells from rheumatoid arthritis patients. Rheumatol Int 2019; 39:819-826. [PMID: 30944956 DOI: 10.1007/s00296-019-04296-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023]
Abstract
We aimed to assess the immunoregulatory effects of secretory factors produced by adipose tissue-derived MSC (AT-MSC) on Th17 and Treg subsets from patients with rheumatoid arthritis (RA). 17 patients with active disease matching the ACR/EULAR 2010 criteria for RA were included. Patients' peripheral blood mononuclear cells (PBMC) were cultured in AT-MSC-conditioned medium (AT-MSCcm) and in control medium. The cytokine production of AT-MSC and PBMC was quantified by ELISA. Th17 and Treg were determined by flow cytometry. AT-MSCcm contained: IL-6, IL-17, IL-21, CCL2, CCL5, IL-8, sVEGF-A and PGE2. Cultivation of patients' PBMC with AT-MSCcm increased TGF-β1 (8318 pg/ml; IQR 6327-11,686) vs control medium [6227 pg/ml (IQR 1681-10,148, p = 0.013)]. PBMC cultivated with AT-MSCcm downregulated TNF-α, IL-17A, and IL-21 compared to control PBMC: 5 pg/ml IQR (1.75-11.65) vs 1 pg/ml (IQR 0.7-1.9), p = 0.001; 4.2 pg/ml (IQR 3.1-6.1) vs 2.3 pg/ml (IQR.75-5.42), p = 0.017; 66.9 pg/ml (IQR 40.6-107.2) vs 53 pg/ml (IQR 22-73), p = 0.022. Th17 decreased under the influence of AT-MSCcm: 10.13 ± 3.88% vs 8.98 ± 3.58%, p = 0.02. CD4+FoxP3+, CD4+CD25-FoxP3+, and CD4+CD25+FoxP3+ was 11.35 ± 4.1%; 7.13 ± 3.12% and 4.22 ± 2% in control PBMC. Accordingly, CD4+FoxP3+, CD4+CD25-FoxP3+, and CD4+CD25+FoxP3+ significantly increased in PBMC cultured with AT-MSCcm: 15.6 ± 6.1%, p = 0.001; 9.56 ± 5.4%, p = 0.004 and 6.04 ± 3.6%, p = 0.001. All these effects could define MSC-based approaches as adequate avenues for further treatment development in RA.
Collapse
Affiliation(s)
- Georgi Vasilev
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria.
| | - Mariana Ivanova
- Clinic of Rheumatology, University Hospital "St. Ivan Rilski"-Sofia, Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Kalina Tumangelova-Yuzeir
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Ekaterina Krasimirova
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Rumen Stoilov
- Clinic of Rheumatology, University Hospital "St. Ivan Rilski"-Sofia, Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
5
|
Oestbye TKK, Ytteborg E. Preparation and Culturing of Atlantic Salmon Muscle Cells for In Vitro Studies. Methods Mol Biol 2018; 1889:319-330. [PMID: 30367423 DOI: 10.1007/978-1-4939-8897-6_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
This chapter outlines methods for isolating myosatellites from Atlantic salmon (Salmo salar), how to keep them in culture and differentiate them into mature myocytes. The protocol further describes how to trans-differentiate the myocytes into osteoblasts (bone cells).
Collapse
|
6
|
Scaffolds Fabricated from Natural Polymers/Composites by Electrospinning for Bone Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:49-78. [DOI: 10.1007/978-981-13-0950-2_4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
7
|
Samsonraj RM, Paradise CR, Dudakovic A, Sen B, Nair AA, Dietz AB, Deyle DR, Cool SM, Rubin J, van Wijnen AJ. Validation of Osteogenic Properties of Cytochalasin D by High-Resolution RNA-Sequencing in Mesenchymal Stem Cells Derived from Bone Marrow and Adipose Tissues. Stem Cells Dev 2018; 27:1136-1145. [PMID: 29882479 DOI: 10.1089/scd.2018.0037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Differentiation of mesenchymal stromal/stem cells (MSCs) involves a series of molecular signals and gene transcription events required for attaining cell lineage commitment. Modulation of the actin cytoskeleton using cytochalasin D (CytoD) drives osteogenesis at early timepoints in bone marrow-derived MSCs and also initiates a robust osteogenic differentiation program in adipose tissue-derived MSCs. To understand the molecular basis for these pronounced effects on osteogenic differentiation, we investigated global changes in gene expression in CytoD-treated murine and human MSCs by high-resolution RNA-sequencing (RNA-seq) analysis. A three-way bioinformatic comparison between human adipose tissue-derived MSCs (hAMSCs), human bone marrow-derived MSCs (hBMSCs), and mouse bone marrow-derived MSCs (mBMSCs) revealed significant upregulation of genes linked to extracellular matrix organization, cell adhesion and bone metabolism. As anticipated, the activation of these differentiation-related genes is accompanied by a downregulation of nuclear and cell cycle-related genes presumably reflecting cytostatic effects of CytoD. We also identified eight novel CytoD activated genes-VGLL4, ARHGAP24, KLHL24, RCBTB2, BDH2, SCARF2, ACAD10, HEPH-which are commonly upregulated across the two species and tissue sources of our MSC samples. We selected the Hippo pathway-related VGLL4 gene, which encodes the transcriptional co-factor Vestigial-like 4, for further study because this pathway is linked to osteogenesis. VGLL4 small interfering RNA depletion reduces mineralization of hAMSCs during CytoD-induced osteogenic differentiation. Together, our RNA-seq analyses suggest that while the stimulatory effects of CytoD on osteogenesis are pleiotropic and depend on the biological state of the cell type, a small group of genes including VGLL4 may contribute to MSC commitment toward the bone lineage.
Collapse
Affiliation(s)
| | - Christopher R Paradise
- 2 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences , Mayo Clinic, Rochester, Minnesota.,3 Center for Regenerative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Amel Dudakovic
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Buer Sen
- 4 Department of Medicine, University of North Carolina , Chapel Hill, North Carolina
| | - Asha A Nair
- 5 Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic , Rochester, Minnesota
| | - Allan B Dietz
- 6 Laboratory Medicine and Pathology, Mayo Clinic , Rochester, Minnesota
| | - David R Deyle
- 7 Department of Medical Genetics, Mayo Clinic , Rochester, Minnesota
| | - Simon M Cool
- 8 Glycotherapeutics Group, Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Janet Rubin
- 3 Center for Regenerative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Andre J van Wijnen
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota.,3 Center for Regenerative Medicine, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
8
|
Current Perspectives Regarding Stem Cell-Based Therapy for Liver Cirrhosis. Can J Gastroenterol Hepatol 2018; 2018:4197857. [PMID: 29670867 PMCID: PMC5833156 DOI: 10.1155/2018/4197857] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
Liver cirrhosis is a major cause of mortality and a common end of various progressive liver diseases. Since the effective treatment is currently limited to liver transplantation, stem cell-based therapy as an alternative has attracted interest due to promising results from preclinical and clinical studies. However, there is still much to be understood regarding the precise mechanisms of action. A number of stem cells from different origins have been employed for hepatic regeneration with different degrees of success. The present review presents a synopsis of stem cell research for the treatment of patients with liver cirrhosis according to the stem cell type. Clinical trials to date are summarized briefly. Finally, issues to be resolved and future perspectives are discussed with regard to clinical applications.
Collapse
|
9
|
Challenges for Cartilage Regeneration. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2017. [DOI: 10.1007/978-3-662-53574-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
10
|
Maerz JK, Roncoroni LP, Goldeck D, Abruzzese T, Kalbacher H, Rolauffs B, DeZwart P, Nieselt K, Hart ML, Klein G, Aicher WK. Bone marrow-derived mesenchymal stromal cells differ in their attachment to fibronectin-derived peptides from term placenta-derived mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:29. [PMID: 26869043 PMCID: PMC4751672 DOI: 10.1186/s13287-015-0243-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/03/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Human mesenchymal stromal cells (MSCs) can be isolated from different sources including bone marrow and term placenta. These two populations display distinct patterns of proliferation and differentiation in vitro. Since proliferation and differentiation of cells are modulated by cell-matrix interactions, we investigated the attachment of MSCs to a set of peptide-coated surfaces and explored their interactions with peptides in suspension. METHODS Human MSCs were isolated from bone marrow and term placenta and expanded. Binding of MSCs to peptides was investigated by a cell-attachment spot assay, by blocking experiments and flow cytometry. The integrin expression pattern was explored by a transcript array and corroborated by quantitative reverse transcription polymerase chain reaction and flow cytometry. RESULTS Expanded placenta-derived MSCs (pMSCs) attached well to surfaces coated with fibronectin-derived peptides P7, P15, and P17, whereas bone marrow-derived MSCs (bmMSCs) attached to P7, but barely to P15 and P17. The binding of bmMSCs and pMSCs to the peptides was mediated by β1 integrins. In suspension, expanded bmMSCs barely bind to P7, P13, P15, and less to P14 and P17. Ex vivo, bmMSCs failed to bind P7, but displayed a weak interaction with P13, P14, and P15. In suspension, expanded pMSCs displayed binding to many peptides, including P4, P7, P13, P14, P15, and P17. The differences observed in binding of bmMSCs and pMSCs to the peptides were associated with significant differences in expression of integrin α2-, α4-, and α6-chains. CONCLUSIONS Human bmMSCs and pMSCs show distinct patterns of attachment to defined peptides and maintain differences in expression of integrins in vitro. Interactions of ex vivo bmMSCs with a given peptide yield different staining patterns compared to expanded bmMSCs in suspension. Attachment of expanded MSCs to peptides on surfaces is different from interactions of expanded MSCs with peptides in suspension. Studies designed to investigate the interactions of human MSCs with peptide-augmented scaffolds or peptides in suspension must therefore regard these differences in cell-peptide interactions.
Collapse
Affiliation(s)
- Jan K Maerz
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Lorenzo P Roncoroni
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - David Goldeck
- Center for Medical Research, Department of Medicine II, University of Tübingen, Tübingen, Germany.
| | - Tanja Abruzzese
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| | - Bernd Rolauffs
- BG Trauma Center Tübingen, University of Tübingen, Tübingen, Germany.
| | - Peter DeZwart
- BG Trauma Center Tübingen, University of Tübingen, Tübingen, Germany.
| | - Kay Nieselt
- Integrative Transcriptomics, Center for Bioinformatics, University of Tübingen, Tübingen, Germany.
| | - Melanie L Hart
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| | - Gerd Klein
- Center for Medical Research, Department of Medicine II, University of Tübingen, Tübingen, Germany.
| | - Wilhelm K Aicher
- KFO273, Department of Urology, University of Tübingen Hospital, Paul Ehrlich Str. 15, 72076, Tübingen, Germany.
| |
Collapse
|
11
|
Rakian R, Block TJ, Johnson SM, Marinkovic M, Wu J, Dai Q, Dean DD, Chen XD. Native extracellular matrix preserves mesenchymal stem cell "stemness" and differentiation potential under serum-free culture conditions. Stem Cell Res Ther 2015; 6:235. [PMID: 26620283 PMCID: PMC4666167 DOI: 10.1186/s13287-015-0235-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/10/2015] [Accepted: 11/10/2015] [Indexed: 02/07/2023] Open
Abstract
Introduction Bone marrow-derived mesenchymal stem cells (BM-MSCs) for clinical use should not be grown in media containing fetal bovine serum (FBS), because of serum-related concerns over biosafety and batch-to-batch variability. Previously, we described the preparation and use of a cell-free native extracellular matrix (ECM) made by bone marrow cells (BM-ECM) which preserves stem cell properties and enhances proliferation. Here, we compare colony-forming ability and differentiation of MSCs cultured on BM-ECM with a commercially available matrix (CELLstart™) and tissue culture plastic (TCP) under serum-free conditions. Methods Primary MSCs from freshly isolated bone marrow-derived mononuclear cells or passaged MSCs (P1) were grown in serum-containing (SCM) or serum-free (SFM) media on BM-ECM, CELLstart™, or TCP substrates. Proliferation, cell composition (phenotype), colony-forming unit replication, and bone morphogenetic protein-2 (BMP-2) responsiveness were compared among cells maintained on the three substrates. Results Proliferation of primary BM-MSCs was significantly higher in SCM than SFM, irrespectively of culture substrate, suggesting that the expansion of these cells requires SCM. In contrast, passaged cells cultured on BM-ECM or CELLstart™ in SFM proliferated to nearly the same extent as cells in SCM. However, morphologically, those on BM-ECM were smaller and more aligned, slender, and long. Cells grown for 7 days on BM-ECM in SFM were 20–40 % more positive for MSC surface markers than cells cultured on CELLstart™. Cells cultured on TCP contained the smallest number of cells positive for MSC markers. MSC colony-forming ability in SFM, as measured by CFU-fibroblasts, was increased 10-, 9-, and 2-fold when P1 cells were cultured on BM-ECM, CELLstart™, and TCP, respectively. Significantly, CFU-adipocyte and -osteoblast replication of cells grown on BM-ECM was dramatically increased over those on CELLstart™ (2X) and TCP (4-7X). BM-MSCs, cultured in SFM and treated with BMP-2, retained their differentiation capacity better on BM-ECM than on either of the other two substrates. Conclusions Our findings indicate that BM-ECM provides a unique microenvironment that supports the colony-forming ability of MSCs in SFM and preserves their stem cell properties. The establishment of a robust culture system, combining native tissue-specific ECM and SFM, provides an avenue for preparing significant numbers of potent MSCs for cell-based therapies in patients.
Collapse
Affiliation(s)
- Rubie Rakian
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - Travis J Block
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - Shannan M Johnson
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA. .,Periodontics Graduate Program, Wilford Hall 59th Medical Wing, 2133 Pepperrell Street, Building 3352, Joint Base San Antonio, Lackland, TX, 78236, USA.
| | - Milos Marinkovic
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - Junjie Wu
- Department of Orthodontics, Fourth Military Medical University, School of Stomatology, 145 West Chang-le Road, Xi'an, Shaanxi Province, 710032, P.R. China.
| | - Qiuxia Dai
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - David D Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA. .,Research Service, Audie Murphy VA Medical Center, South Texas Veterans Health Care System, 7400 Merton Minter Boulevard, San Antonio, TX, 78229-4404, USA.
| |
Collapse
|
12
|
Ke X, Li Q, Xu L, Zhang Y, Li D, Ma J, Mao X. Netrin-1 overexpression in bone marrow mesenchymal stem cells promotes functional recovery in a rat model of peripheral nerve injury. J Biomed Res 2015; 29:380-389. [PMID: 26445571 PMCID: PMC4585432 DOI: 10.7555/jbr.29.20140076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 10/01/2014] [Accepted: 04/08/2015] [Indexed: 11/08/2022] Open
Abstract
Transplantation of bone marrow mesenchymal stem cells (BMSCs) has been developed as a new method of treating diseases of the peripheral nervous system. While netrin-1 is a critical molecule for axonal path finding and nerve growth, it may also affect vascular network formation. Here, we investigated the effect of transplanting BMSCs that produce netrin-1 in a rat model of sciatic nerve crush injury. We introduced a sciatic nerve crush injury, and then injected 1×10(6) BMSCs infected by a recombinant adenovirus expressing netrin-1 Ad5-Netrin-1-EGFP or culture medium into the injured part in the next day. At day 7, 14 and 28 after injection, we measured motor nerve conduction and detected mRNA expressions of netrin-1 receptors UNC5B and Deleted in Colorectal Cancer (DCC), and neurotrophic factors brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) by real-time PCR. We also detected protein expressions of BDNF and NGF by Western blotting assays and examined BMSCs that incorporated into myelin and vascellum. The results showed that BMSCs infected by Ad5-Netrin-1-EGFP significantly improved the function of the sciatic nerve, and led to increased expression of BDNF and NGF (P<0.05). Moreover, 28 days after injury, more Schwann cells were found in BMSCs infected by Ad5-Netrin-1-EGFP compared to control BMSCs. In conclusion, transplantation of BMSCs that produce netrin-1 improved the function of the sciatic nerve after injury. This method may be a new treatment of nerve injury.
Collapse
Affiliation(s)
- Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Li Xu
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Ying Zhang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Dongmei Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Xiaoming Mao
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| |
Collapse
|
13
|
Ulrich C, Abruzzese T, Maerz JK, Ruh M, Amend B, Benz K, Rolauffs B, Abele H, Hart ML, Aicher WK. Human Placenta-Derived CD146-Positive Mesenchymal Stromal Cells Display a Distinct Osteogenic Differentiation Potential. Stem Cells Dev 2015; 24:1558-69. [DOI: 10.1089/scd.2014.0465] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Christine Ulrich
- Center for Regenerative Medicine, University of Tübingen Hospital, Tübingen, Germany
| | - Tanja Abruzzese
- Department of Urology, University of Tübingen Hospital, Tübingen, Germany
| | - Jan K. Maerz
- Center for Regenerative Medicine, University of Tübingen Hospital, Tübingen, Germany
| | - Manuel Ruh
- Center for Regenerative Medicine, University of Tübingen Hospital, Tübingen, Germany
| | - Bastian Amend
- Department of Urology, University of Tübingen Hospital, Tübingen, Germany
| | - Karin Benz
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Bernd Rolauffs
- Department of Traumatology, BGU Hospital, University of Tübingen, Tübingen, Germany
| | - Harald Abele
- Department of Gynecology and Obstetrics, University of Tübingen Hospital, Tübingen, Germany
| | - Melanie L. Hart
- Department of Urology, University of Tübingen Hospital, Tübingen, Germany
| | - Wilhelm K. Aicher
- Center for Regenerative Medicine, University of Tübingen Hospital, Tübingen, Germany
- Department of Urology, University of Tübingen Hospital, Tübingen, Germany
| |
Collapse
|
14
|
Ytteborg E, Todorcevic M, Krasnov A, Takle H, Kristiansen IØ, Ruyter B. Precursor cells from Atlantic salmon (Salmo salar) visceral fat holds the plasticity to differentiate into the osteogenic lineage. Biol Open 2015; 4:783-91. [PMID: 25948755 PMCID: PMC4571100 DOI: 10.1242/bio.201411338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In order to study the potential plasticity of Atlantic salmon (Salmo salar) precursor cells (aSPCs) from the adipogenic mesenchyme cell lineage to differentiate to the osteogenic lineage, aSPCs were isolated and cultivated under either osteogenic or adipogenic promoting conditions. The results strengthen the hypothesis that aSPCs most likely are predestined to the adipogenic lineage, but they also hold the flexibility to turn into other lineages given the right stimuli. This assumption is supported by the fact that the transcription factor pparγ , important for regulation of adiopogenesis, was silent in aSPCs grown in osteogenic media, while runx2, important for osteogenic differentiation, was not expressed in aSPCs cultivated in adipogenic media. After 2 weeks in osteogenic promoting conditions the cells started to deposit extracellular matrix and after 4 weeks, the cells started mineralizing secreted matrix. Microarray analyses revealed large-scale transcriptome responses to osteogenic medium after 2 days, changes remained stable at day 15 and decreased by magnitude at day 30. Induction was observed in many genes involved in osteogenic differentiation, growth factors, regulators of development, transporters and production of extracellular matrix. Transcriptome profile in differentiating adipocytes was markedly different from differentiating osteoblasts with far fewer genes changing activity. The number of regulated genes slowly increased at the mature stage, when adipocytes increased in size and accumulated lipids. This is the first report on in vitro differentiation of aSPCs from Atlantic salmon to mineralizing osteogenic cells. This cell model system provides a new valuable tool for studying osteoblastogenesis in fish.
Collapse
|
15
|
Naqvi SM, Buckley CT. Differential Response of Encapsulated Nucleus Pulposus and Bone Marrow Stem Cells in Isolation and Coculture in Alginate and Chitosan Hydrogels. Tissue Eng Part A 2015; 21:288-99. [DOI: 10.1089/ten.tea.2013.0719] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Syeda Masooma Naqvi
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Conor Timothy Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Vainshtein JM, Kabarriti R, Mehta KJ, Roy-Chowdhury J, Guha C. Bone marrow-derived stromal cell therapy in cirrhosis: clinical evidence, cellular mechanisms, and implications for the treatment of hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 2014; 89:786-803. [PMID: 24969793 DOI: 10.1016/j.ijrobp.2014.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 01/18/2023]
Abstract
Current treatment options for hepatocellular carcinoma (HCC) are often limited by the presence of underlying liver disease. In patients with liver cirrhosis, surgery, chemotherapy, and radiation therapy all carry a high risk of hepatic complications, ranging from ascites to fulminant liver failure. For patients receiving radiation therapy, cirrhosis dramatically reduces the already limited radiation tolerance of the liver and represents the most important clinical risk factor for the development of radiation-induced liver disease. Although improvements in conformal radiation delivery techniques have improved our ability to safely irradiate confined areas of the liver to increasingly higher doses with excellent local disease control, patients with moderate-to-severe liver cirrhosis continue to face a shortage of treatment options for HCC. In recent years, evidence has emerged supporting the use of bone marrow-derived stromal cells (BMSCs) as a promising treatment for liver cirrhosis, with several clinical studies demonstrating sustained improvement in clinical parameters of liver function after autologous BMSC infusion. Three predominant populations of BMSCs, namely hematopoietic stem cells, mesenchymal stem cells, and endothelial progenitor cells, seem to have therapeutic potential in liver injury and cirrhosis. Preclinical studies of BMSC transplantation have identified a range of mechanisms through which these cells mediate their therapeutic effects, including hepatocyte transdifferentiation and fusion, paracrine stimulation of hepatocyte proliferation, inhibition of activated hepatic stellate cells, enhancement of fibrolytic matrix metalloproteinase activity, and neovascularization of regenerating liver. By bolstering liver function in patients with underlying Child's B or C cirrhosis, autologous BMSC infusion holds great promise as a therapy to improve the safety, efficacy, and utility of surgery, chemotherapy, and hepatic radiation therapy in the treatment of HCC.
Collapse
Affiliation(s)
| | - Rafi Kabarriti
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Keyur J Mehta
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jayanta Roy-Chowdhury
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Genetics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
17
|
Ngo MA, Müller A, Li Y, Neumann S, Tian G, Dixon IMC, Arora RC, Freed DH. Human mesenchymal stem cells express a myofibroblastic phenotype in vitro: comparison to human cardiac myofibroblasts. Mol Cell Biochem 2014; 392:187-204. [PMID: 24691634 DOI: 10.1007/s11010-014-2030-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 03/14/2014] [Indexed: 01/12/2023]
Abstract
Cardiac fibrosis accompanies a variety of myocardial disorders, and is induced by myofibroblasts. These cells may be composed of a heterogeneous population of parent cells, including interstitial fibroblasts and circulating progenitor cells. Direct comparison of human bone marrow-derived mesenchymal stem cells (BM-MSCs) and cardiac myofibroblasts (CMyfbs) has not been previously reported. We hypothesized that BM-MSCs readily adopt a myofibroblastic phenotype in culture. Human primary BM-MSCs and human CMyfbs were isolated from patients undergoing open heart surgery and expanded under standard culture conditions. We assessed and compared their phenotypic and functional characteristics by examining their gene expression profile, their ability to contract collagen gels and synthesize collagen type I. In addition, we examined the role of non-muscle myosin II (NMMII) in modulating MSC myogenic function using NMMII siRNA knockdown and blebbistatin, a specific small molecule inhibitor of NMMII. We report that, while human BM-MSCs retain pluripotency, they adopt a myofibroblastic phenotype in culture and stain positive for the myofibroblast markers α-SMA, vimentin, NMMIIB, ED-A fibronectin, and collagen type 1 at each passage. In addition, they contract collagen gels in response to TGF-β1 and synthesize collagen similar to human CMyfbs. Moreover, inhibition of NMMII activity with blebbistatin completely attenuates gel contractility without affecting cell viability. Thus, human BM-MSCs share and exhibit similar physiological and functional characteristics as human CMyfbs in vitro, and their propensity to adopt a myofibroblast phenotype in culture may contribute to cardiac fibrosis.
Collapse
Affiliation(s)
- Melanie A Ngo
- Department of Physiology, Faculty of Medicine, Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang A, Midura RJ, Vasanji A, Wang AJ, Hascall VC. Hyperglycemia diverts dividing osteoblastic precursor cells to an adipogenic pathway and induces synthesis of a hyaluronan matrix that is adhesive for monocytes. J Biol Chem 2014; 289:11410-11420. [PMID: 24569987 DOI: 10.1074/jbc.m113.541458] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Isolated rat bone marrow stromal cells cultured in osteogenic medium in which the normal 5.6 mm glucose is changed to hyperglycemic 25.6 mm glucose greatly increase lipid formation between 21-31 days of culture that is associated with decreased biomineralization, up-regulate expression of cyclin D3 and two adipogenic markers (CCAAT/enhancer binding protein α and peroxisome proliferator-activated receptor γ) within 5 days of culture, increase neutral and polar lipid synthesis within 5 days of culture, and form a monocyte-adhesive hyaluronan matrix through an endoplasmic reticulum stress-induced autophagic mechanism. Evidence is also provided that, by 4 weeks after diabetes onset in the streptozotocin-induced diabetic rat model, there is a large loss of trabecular bone mineral density without apparent proportional changes in underlying collagen matrices, a large accumulation of a hyaluronan matrix within the trabecular bone marrow, and adipocytes and macrophages embedded in this hyaluronan matrix. These results support the hypothesis that hyperglycemia in bone marrow diverts dividing osteoblastic precursor cells (bone marrow stromal cells) to a metabolically stressed adipogenic pathway that induces synthesis of a hyaluronan matrix that recruits inflammatory cells and establishes a chronic inflammatory process that demineralizes trabecular cancellous bone.
Collapse
Affiliation(s)
- Aimin Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195.
| | - Ronald J Midura
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Amit Vasanji
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Andrew J Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
19
|
Melchiorri AJ, Nguyen BNB, Fisher JP. Mesenchymal stem cells: roles and relationships in vascularization. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:218-28. [PMID: 24410463 DOI: 10.1089/ten.teb.2013.0541] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
One of the primary challenges in translating tissue engineering to clinical applicability is adequate, functional vascularization of tissue constructs. Vascularization is necessary for the long-term viability of implanted tissue expanded and differentiated in vitro. Such tissues may be derived from various cell sources, including mesenchymal stem cells (MSCs). MSCs, able to differentiate down several lineages, have been extensively researched for their therapeutic capabilities. In addition, MSCs have a variety of roles in the vascularization of tissue, both through direct contact and indirect signaling. The studied relationships between MSCs and vascularization have been utilized to further the necessary advancement of vascularization in tissue engineering concepts. This review aims to provide a summary of relevant relationships between MSCs, vascularization, and other relevant cell types, along with an overview discussing applications and challenges related to the roles and relationships of MSCs and vascular tissues.
Collapse
Affiliation(s)
- Anthony J Melchiorri
- Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | | | | |
Collapse
|
20
|
Skurikhin EG, Khmelevskaya ES, Pershina OV, Ermakova NN, Krupin VA, Reztsova AM, Ermolaeva LA, Yakushina VD, Stepanova IE, Reztsova VM, Cherdyntseva NV, Stakheeva MN, Dygai AM. Differentiation of mesenchymal multipotent stromal cells of the lungs in pneumofibrosis. Bull Exp Biol Med 2013; 154:537-43. [PMID: 23486599 DOI: 10.1007/s10517-013-1995-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We studied differentiation of multipotent mesenchymal stromal cells (MMSC) of the lungs of C57Bl/6 mice with bleomycin-induced pneumofibrosis. Adherent mononuclear cells found in mouse lungs demonstrated mesenchymal phenotype and expressed CD44, CD73, CD90, and CD106, but not CD31, CD34, and CD45. The cells with MMSC characteristics differentiate in vitro into various cells of stromal lines (chondrocytes, osteogenic cells, adipocytes, and fibroblasts). Bleomycin increased the growth rate of MMSC and selectively promoted their differentiation towards fibroblast cells.
Collapse
Affiliation(s)
- E G Skurikhin
- Research Institute of Pharmacology, Siberian Division of the Russian Academy of Medical Sciences, Tomsk, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pekovits K, Kröpfl JM, Stelzer I, Payer M, Hutter H, Dohr G. Human mesenchymal progenitor cells derived from alveolar bone and human bone marrow stromal cells: a comparative study. Histochem Cell Biol 2013; 140:611-21. [PMID: 23996194 DOI: 10.1007/s00418-013-1140-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2013] [Indexed: 01/09/2023]
Abstract
The aim of the present study was to evaluate the potential of intraoral harvested alveolar bone as an alternative source of multipotent mesenchymal stromal cells for future applications in oral and maxillofacial tissue engineering. Explant cultures were established from 20 alveolar bone samples harvested from the oblique line immediately before wisdom tooth removal. Morphology and proliferation characteristics of the in vitro expanded cells, referred to as human alveolar bone-derived cells (hABDCs), were studied using phase-contrast microscopy. Immunocytochemical analysis of their surface marker expression was conducted using monoclonal antibodies defining mesenchymal stromal cells. To evaluate their multilineage differentiation potential, hABDCs were induced to differentiate along the osteogenic, adipogenic, and chondrogenic lineage and compared to bone marrow mesenchymal stromal cells (hBMSCs) on mRNA and protein levels applying RT-PCR and cytochemical staining methods. hABDCs showed typical morphological characteristics comparable to those of hBMSCs such as being mononuclear, fibroblast-like, spindle-shaped, and plastic adherent. Immunophenotypically, cells were positive for CD105, CD90, and CD73 while negative for CD45, CD34, CD14, CD79α, and HLA-DR surface molecules, indicating an antigen expression pattern considered typical for multipotent mesenchymal stromal cells. As evidenced by RT-PCR and cytochemistry, hABDCs showed multilineage differentiation and similar chondrogenic and osteogenic differentiation potentials when compared to hBMSCs. Our findings demonstrate that human alveolar bone contains mesenchymal progenitor cells that can be isolated and expanded in vitro and are capable of trilineage differentiation, providing a reservoir of multipotent mesenchymal cells from an easily accessible tissue source.
Collapse
Affiliation(s)
- Karin Pekovits
- Institute of Cell Biology, Histology and Embryology, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/7, 8010, Graz, Austria,
| | | | | | | | | | | |
Collapse
|
22
|
Ulrich C, Rolauffs B, Abele H, Bonin M, Nieselt K, Hart ML, Aicher WK. Low osteogenic differentiation potential of placenta-derived mesenchymal stromal cells correlates with low expression of the transcription factors Runx2 and Twist2. Stem Cells Dev 2013; 22:2859-72. [PMID: 23763516 DOI: 10.1089/scd.2012.0693] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recent studies indicated that mesenchymal stromal cells from bone marrow (bmMSC) differ in their osteogenic differentiation capacity compared to MSC from term placenta (pMSC). We extended these studies and investigated the expression of factors involved in regulation of bone metabolism in both cell types. To this end, MSC were expanded in vitro and characterized. The total transcriptome was investigated by microarrays, and for selected genes, the differences in gene expression were explored by quantitative reverse transcriptase-polymerase chain reaction, immunocytochemistry, and flow cytometry. We report that bmMSC and pMSC share expression of typical lineage surface markers, including CD73, CD90, CD105, and lack of CD14, CD34, and CD45. However, according to transcriptome analyses, they differ significantly in their expression of more than 590 genes. Factors involved in bone metabolism, including alkaline phosphatase (P<0.05), osteoglycin (P<0.05), osteomodulin (P<0.05), runt-related transcription factor 2 (Runx2) (P<0.04), and WISP2 (P<0.05), were expressed at significantly lower levels in pMSC, but twist-related protein 2 (Twist2) (P<0.0002) was expressed at significantly higher levels. The osteogenic differentiation capacity of pMSC was very low. The adipogenic differentiation was somewhat more prominent in bmMSC, while the chondrogenic differentiation seemed not to differ between bmMSC and pMSC, as determined by histochemical staining. However, expression and induction of peroxisome proliferator-activated receptor gamma-2 (PPARγ2) and Sox9, factors involved in early adipogenesis and chondrogenesis, respectively, were higher in bmMSC. We conclude that despite many similarities between bmMSC and pMSC, when expanded under identical conditions, they vary considerably with respect to their in vitro differentiation potential. For regenerative purposes, the choice of MSC may therefore influence the outcome of a treatment considerably.
Collapse
Affiliation(s)
- Christine Ulrich
- 1 ZRM, Center for Regenerative Medicine, University of Tuebingen , Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Czernik M, Fidanza A, Sardi M, Galli C, Brunetti D, Malatesta D, Della Salda L, Matsukawa K, Ptak GE, Loi P. Differentiation potential and GFP labeling of sheep bone marrow-derived mesenchymal stem cells. J Cell Biochem 2013; 114:134-43. [PMID: 22886939 DOI: 10.1002/jcb.24310] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 07/26/2012] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) are an important cell population in the bone marrow microenvironment. MSCs have the capacity to differentiate in vitro into several mesenchymal tissues including bone, cartilage, fat, tendon, muscle, and marrow stroma. This study was designed to isolate, expand, and characterize the differentiation ability of sheep bone marrow-derived MSCs and to demonstrate the possibility to permanently express a reporter gene. Bone marrow was collected from the iliac crest and mononuclear cells were separated by density gradient centrifugation. Sheep MSCs cell lines were stable characterized as CD44+ and CD34- and then transfected with a green fluorescent protein (GFP) reporter gene. The GFP expression was maintained in about half (46.6%) of cloned blastocysts produced by nuclear transfer of GFP+ sheep MSCs, suggesting the possibility to establish multipotent embryonic cells' lines carrying the fluorescent tag for comparative studies on the differentiation capacity of adult stem cells (MSCs) versus embryonic stem cells. We found that sheep MSCs under appropriate culture conditions could be induced to differentiate into adipocytes, chondrocytes, and osteoblast lineages. Our results confirm the plasticity of sheep MSCs and establish the foundation for the development of a pre-clinical sheep model to test the efficiency and safety of cell replacement therapy.
Collapse
Affiliation(s)
- Marta Czernik
- Department of Comparative Biomedical Science, University of Teramo, 64100 Teramo, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Luo D, Yan X, Liu D, Zhou X, Liu G. Differential effects of mesenchymal stem cells on a heterogeneous cell population within lung cancer cell lines. Mol Cell Biochem 2013; 378:107-16. [PMID: 23456479 DOI: 10.1007/s11010-013-1600-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/23/2013] [Indexed: 12/19/2022]
Abstract
Although mesenchymal stem cells (MSCs) promote lung cancer growth in vivo, in vitro studies indicate that they inhibit the proliferation of lung cancer cells. Because malignant tumors contain a heterogeneous cell population with variable capacity for self-renewal, the aim of this study was to determine whether the inconsistencies between in vitro and in vivo studies are a result of differential effects of MSCs on the heterogeneous cell population within lung cancer cell lines. Human MSCs were isolated from the bone marrow, and their cell surface antigen expression and multi-lineage differentiation capacity was examined at passage 10. CD133+ cells were isolated from A549 and H446 cell lines using immunomagnetic separation. The effects of MSCs on the growth and microsphere formation of heterogeneous cell populations within two lung cancer cell lines (A549 and H446) were compared. MSCs inhibited the in vitro proliferation of both cell lines, but significantly accelerated tumor formation and stimulated tumor growth in vivo (P < 0.05). In CD133+ cells isolated from both A549 and H446 cells, co-culture with MSCs for 1-3 days significantly increased their proliferation (P < 0.05). MSCs also significantly increased microsphere formation in both cell lines (P < 0.05). Selective stimulation of CD133+ cell growth may account for the discrepant effects of MSCs on lung cancer progression.
Collapse
Affiliation(s)
- Dan Luo
- Department of Respiratory, Southwest Hospital, Third Military Medical University of PLA, Shapingba District, Chongqing, China
| | | | | | | | | |
Collapse
|
25
|
Assessing adipogenic potential of mesenchymal stem cells: a rapid three-dimensional culture screening technique. Stem Cells Int 2013; 2013:806525. [PMID: 23431315 PMCID: PMC3574742 DOI: 10.1155/2013/806525] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/07/2012] [Accepted: 12/26/2012] [Indexed: 01/11/2023] Open
Abstract
Bone-marrow-derived mesenchymal stem cells (MSCs) have the potential to differentiate into a number of phenotypes, including adipocytes. Adipogenic differentiation has traditionally been performed in monolayer culture, and, while the expression of a fat-cell phenotype can be achieved, this culture method is labor and material intensive and results in only small numbers of fragile adherent cells, which are not very useful for further applications. Aggregate culture is a cell-culture technique in which cells are induced to form three-dimensional aggregates; this method has previously been used successfully, among others, to induce and study chondrogenic differentiation of MSCs. We have previously published an adaptation of the chondrogenic aggregate culture method to a 96-well plate format. Based on the success of this method, we have used the same format for the preparation of three-dimensional adipogenic cultures. The MSCs differentiate rapidly, the aggregates can be handled and processed for histologic and biochemical assays with ease, and the format offers significant savings in supplies and labor. As a differentiation assay, this method can distinguish between degrees of senescence and appears suitable for testing medium or drug formulations in a high-volume, high-throughput fashion.
Collapse
|
26
|
Abstract
Stem cell transplantation is a promising approach for improving cardiac function after severe myocardial damage for which use of autologous cells have been preferred to avoid immune rejection. Recently, however, rodent as well as human mesenchymal stromal cells (MSCs) have been reported to be uniquely immune tolerant, both in in vitro as well as in vivo transplant models. In this chapter, we summarize the current understanding of the underlying immunologic mechanisms, which can facilitate the use of such cells as "universal donor cells."
Collapse
|
27
|
Webber MJ, Matson JB, Tamboli VK, Stupp SI. Controlled release of dexamethasone from peptide nanofiber gels to modulate inflammatory response. Biomaterials 2012; 33:6823-32. [PMID: 22748768 PMCID: PMC3445268 DOI: 10.1016/j.biomaterials.2012.06.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/02/2012] [Indexed: 01/17/2023]
Abstract
New biomaterials that have the ability to locally suppress an immune response could have broad therapeutic use in the treatment of diseases characterized by acute or chronic inflammation or as a strategy to facilitate improved efficacy in cell or tissue transplantation. We report here on the preparation of a modular peptide amphiphile (PA) capable of releasing an anti-inflammatory drug, dexamethasone (Dex), by conjugation via a labile hydrazone linkage. This molecule self-assembled in water into long supramolecular nanofibers when mixed with a similar PA lacking the drug conjugate, and the addition of calcium salt to screen electrostatic repulsion between nanofibers promoted gel formation. These nanofiber gels demonstrated sustained release of soluble Dex for over one month in physiologic media. The Dex released from these gels maintained its anti-inflammatory activity when evaluated in vitro using a human inflammatory reporter cell line and furthermore preserved cardiomyocyte viability upon induced oxidative stress. The ability of this gel to mitigate the inflammatory response in cell transplantation strategies was evaluated using cell-surrogate polystyrene microparticles suspended in the nanofiber gel that were then subcutaneously injected into mice. Live animal luminescence imaging using the chemiluminescent reporter molecule luminol showed a significant reduction in inflammation at the site where particles were injected with Dex-PA compared to the site of injection for particles within a control PA in the same animal. Histological evidence suggested a marked reduction in the number of infiltrating inflammatory cells when particles were delivered within Dex-PA nanofiber gels, and very little inflammation was observed at either 3 days or 21 days post-implantation. The use of Dex-PA could facilitate localized anti-inflammatory activity as a component of biomaterials designed for various applications in regenerative medicine and could specifically be a useful module for PA-based therapies. More broadly, these studies define a versatile strategy for facile synthesis of self-assembling peptide-based materials with the ability to control drug release.
Collapse
Affiliation(s)
- Matthew J. Webber
- Northwestern University, Biomedical Engineering Department, Evanston, IL 60208
| | - John B. Matson
- Northwestern University, Institute for BioNanotechnology in Medicine, Chicago, IL 60611
| | - Vibha K. Tamboli
- Northwestern University, Master of Biotechnology Program, Department of Chemical and Biological Engineering, Evanston, IL 60208
| | - Samuel I. Stupp
- Department of Materials Science and Engineering, Department of Chemistry, Evanston, IL 60208
- Department of Medicine, Institute for BioNanotechnology in Medicine, Chicago, IL 60611
| |
Collapse
|
28
|
Cristante AF, Barros Filho TEPD, Marcon RM, Letaif OB, Rocha IDD. Therapeutic approaches for spinal cord injury. Clinics (Sao Paulo) 2012; 67:1219-24. [PMID: 23070351 PMCID: PMC3460027 DOI: 10.6061/clinics/2012(10)16] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 01/02/2023] Open
Abstract
This study reviews the literature concerning possible therapeutic approaches for spinal cord injury. Spinal cord injury is a disabling and irreversible condition that has high economic and social costs. There are both primary and secondary mechanisms of damage to the spinal cord. The primary lesion is the mechanical injury itself. The secondary lesion results from one or more biochemical and cellular processes that are triggered by the primary lesion. The frustration of health professionals in treating a severe spinal cord injury was described in 1700 BC in an Egyptian surgical papyrus that was translated by Edwin Smith; the papyrus reported spinal fractures as a "disease that should not be treated." Over the last biological or pharmacological treatment method. Science is unraveling the mechanisms of cell protection and neuroregeneration, but clinically, we only provide supportive care for patients with spinal cord injuries. By combining these treatments, researchers attempt to enhance the functional recovery of patients with spinal cord injuries. Advances in the last decade have allowed us to encourage the development of experimental studies in the field of spinal cord regeneration. The combination of several therapeutic strategies should, at minimum, allow for partial functional recoveries for these patients, which could improve their quality of life.
Collapse
Affiliation(s)
- Alexandre Fogaça Cristante
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, Instituto de Ortopedia e Traumatologia (IOT), Grupo de Coluna, São Paulo/SP, Brazil
| | | | | | | | | |
Collapse
|
29
|
Abstract
CVD irreversibly damage the cardiomyocytes, the heart muscle cells. This loss triggers a cascade of detrimental events, including formation of scar tissue, an overload of blood flow and pressure capacity, the overstretching of viable cardiac cells, leading to heart failure and eventual death. Restoring damaged heart muscle tissue, through repair or regeneration, is a potentially new strategy to treat heart failure and various other CVD. Stem cells are promising new therapeutics for patients with different heart diseases. The remarkable proliferative and differentiation capacity of stem cells promises an unlimited supply of specific cell types including viable functioning heart muscle cells. A crucial issue in designing more rational cell-based therapy approaches for cardiac disease is understanding the mechanisms by which each of the stem cell or progenitor-cell types can affect myocardial performance. This paper will highlight findings of multiple preliminary clinical experiments involving stem cells as therapeutics, educate the reader on the incidence and prevalence of CVD, the risk factors associated with CVD, and explore some of the challenges that can be encountered.
Collapse
Affiliation(s)
- Amita J Hotkar
- The Milano School of Management and Urban Policy, New School University, 72 Fifth Ave, New York, NY 10011, USA.
| | | |
Collapse
|
30
|
Yu X, Cohen DM, Chen CS. miR-125b Is an adhesion-regulated microRNA that protects mesenchymal stem cells from anoikis. Stem Cells 2012; 30:956-64. [PMID: 22331826 DOI: 10.1002/stem.1064] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have the capacity for multilineage differentiation and are being explored as a source for stem cell-based therapies. Previous studies have shown that adhesion to extracellular matrix plays a critical role in guiding MSC differentiation to distinct lineages. Here, we conducted a focused screen of microRNAs to reveal one microRNA, miR-125b, whose expression changes as a function of cell adhesion. miR-125b expression was upregulated by limiting cell-matrix adhesion using micropatterned substrates, knocking down beta5 integrin or placing cells in suspension culture. Interestingly, we noted that suspending human MSCs (hMSCs) did not induce substantial apoptosis (anoikis) as is typically observed in adherent cells. Although miR-125b appeared to have some effects on hMSC differentiation, we demonstrated a striking role for miR-125b in protecting hMSCs from anoikis. Knockdown of miR-125b increased anoikis while expressing a mimic protected cells. Mechanistic studies demonstrated that miR-125b protected against anoikis by increasing ERK phosphorylation and by suppressing p53. Lastly, we found that miR-125b expression is quite limited in endothelial cells and mouse embryonic fibroblasts (MEFs). The rapid anoikis normally observed in endothelial cells was antagonized by transfection of a miR-125b mimic, suggesting that miR-125b can confer resistance to anoikis in multiple cell types. We also found that endogenous miR-125b was significantly upregulated during reprogramming of MEFs to induced pluripotent cells, suggesting that miR-125b expression may be associated with stem cell populations. Collectively, these observations demonstrate a novel link between cell-matrix adhesion, miR-125b expression, and a stem cell-specific survival program triggered in adhesion-limited contexts such as might occur in early development and wound healing.
Collapse
Affiliation(s)
- Xiang Yu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
31
|
Liu Y, Buckley CT, Downey R, Mulhall KJ, Kelly DJ. The role of environmental factors in regulating the development of cartilaginous grafts engineered using osteoarthritic human infrapatellar fat pad-derived stem cells. Tissue Eng Part A 2012; 18:1531-41. [PMID: 22443147 DOI: 10.1089/ten.tea.2011.0575] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Engineering functional cartilaginous grafts using stem cells isolated from osteoarthritic human tissue is of fundamental importance if autologous tissue engineering strategies are to be used in the treatment of diseased articular cartilage. It has previously been demonstrated that human infrapatellar fat pad (IFP)-derived stem cells undergo chondrogenesis in pellet culture; however, the ability of such cells to generate functional cartilaginous grafts has not been adequately addressed. The objective of this study was to explore how environmental conditions regulate the functional development of cartilaginous constructs engineered using diseased human IFP-derived stem cells (FPSCs). FPSCs were observed to display a diminished chondrogenic potential upon encapsulation in a three-dimensional hydrogel compared with pellet culture, synthesizing significantly lower levels of glycosaminoglycan and collagen on a per cell basis. To engineer more functional cartilaginous grafts, we next explored whether additional biochemical and biophysical stimulations would enhance chondrogenesis within the hydrogels. Serum stimulation was observed to partially recover the diminished chondrogenic potential within hydrogel culture. Over 42 days, stem cells that had first been expanded in a low-oxygen environment proliferated extensively on the outer surface of the hydrogel in response to serum stimulation, assembling a dense type II collagen-positive cartilaginous tissue resembling that formed in pellet culture. The application of hydrostatic pressure did not further enhance extracellular matrix synthesis within the hydrogels, but did appear to alter the spatial accumulation of extracellular matrix leading to the formation of a more compact tissue with superior mechanically functionality. Further work is required in order to recapitulate the environmental conditions present during pellet culture within scaffolds or hydrogels in order to engineer more functional cartilaginous grafts using human osteoarthritic FPSCs.
Collapse
Affiliation(s)
- Yurong Liu
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | |
Collapse
|
32
|
Karantalis V, Balkan W, Schulman IH, Hatzistergos KE, Hare JM. Cell-based therapy for prevention and reversal of myocardial remodeling. Am J Physiol Heart Circ Physiol 2012; 303:H256-70. [PMID: 22636682 DOI: 10.1152/ajpheart.00221.2012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although pharmacological and interventional advances have reduced the morbidity and mortality of ischemic heart disease, there is an ongoing need for novel therapeutic strategies that prevent or reverse progressive ventricular remodeling following myocardial infarction, the process that forms the substrate for ventricular failure. The development of cell-based therapy as a strategy to repair or regenerate injured tissue offers extraordinary promise for a powerful anti-remodeling therapy. In this regard, the field of cell therapy has made major advancements in the past decade. Accumulating data from preclinical studies have provided novel insights into stem cell engraftment, differentiation, and interactions with host cellular elements, as well as the effectiveness of various methods of cell delivery and accuracy of diverse imaging modalities to assess therapeutic efficacy. These findings have in turn guided rationally designed translational clinical investigations. Collectively, there is a growing understanding of the parameters that underlie successful cell-based approaches for improving heart structure and function in ischemic and other cardiomyopathies.
Collapse
Affiliation(s)
- Vasileios Karantalis
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, USA
| | | | | | | | | |
Collapse
|
33
|
Aydemir E, Bayrak OF, Sahin F, Atalay B, Kose GT, Ozen M, Sevli S, Dalan AB, Yalvac ME, Dogruluk T, Türe U. Characterization of cancer stem-like cells in chordoma. J Neurosurg 2012; 116:810-20. [PMID: 22283189 DOI: 10.3171/2011.12.jns11430] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECT Chordomas are locally aggressive bone tumors known to arise from the remnants of the notochord. Because chordomas are rare, molecular studies aimed at developing new therapies are scarce and new approaches are needed. Chordoma cells and cancer stem-like cells share similar characteristics, including self-renewal, differentiation, and resistance to chemotherapy. Therefore, it seems possible that chordomas might contain a subpopulation of cancer stem-like cells. The aim of this study is to determine whether cancer stem-like cells might be present in chordomas. METHODS In this study, the authors used gene expression analysis for common cancer stem-like cellmarkers, including c-myc, SSEA-1, oct4, klf4, sox2, nanog, and brachyury, and compared chordoma cells and tissues with nucleus pulposus tissues (disc degenerated nontumorigenic tissues). Differentiation through agents such as all-trans retinoic acid and osteogenic differentiation medium was induced to the chordoma cells. Additionally, U-CH1 cells were sorted via magnetic cell sorting for stem cell markers CD133 and CD15. After separation, positive and negative cells for these markers were grown in a nonadherent environment, soft agar, to determine whether the presence of these cancer stem-like cells might be responsible for initiating chordoma. The results were compared with those of untreated cells in terms of migration, proliferation, and gene expression by using reverse transcriptase polymerase chain reaction. RESULTS The results indicate that chordoma cells might be differentiating and committing into an osteogenic lineage when induced with the osteogenic differentiation agent. Chordoma cells that are induced with retinoic acid showed slower migration and proliferation rates when compared with the untreated cells. Chordoma cells that were found to be enriched by cancer stem-like cell markers, namely CD133 and CD15, were able to live in a nonadherent soft agar medium, demonstrating a self-renewal capability. To the authors' knowledge, this is the first time that cancer stem-like cell markers were also found to be expressed in chordoma cells and tissues. CONCLUSIONS Cancer stem-like cell detection might be an important step in determining the recurrent and metastatic characteristics of chordoma. This finding may lead to the development of new approaches toward treatments of chordomas.
Collapse
Affiliation(s)
- Esra Aydemir
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kitada M. Mesenchymal cell populations: development of the induction systems for Schwann cells and neuronal cells and finding the unique stem cell population. Anat Sci Int 2012; 87:24-44. [PMID: 22237924 DOI: 10.1007/s12565-011-0128-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 12/20/2011] [Indexed: 01/01/2023]
Abstract
Mesenchymal cell populations, referred to as mesenchymal stem cells or multipotent stromal cells (MSCs), which include bone marrow stromal cells (BMSCs), umbilical cord stromal cells and adipose stromal cells (ASCs), participate in tissue repair when transplanted into damaged or degenerating tissues. The trophic support and immunomodulation provided by MSCs can protect against tissue damage, and the differentiation potential of these cells may help to replace lost cells. MSCs are easily accessible and can be expanded on a large scale. In addition, BMSCs and ASCs can be harvested from the patient himself. Thus, MSCs are considered promising candidates for cell therapy. In this review, I will discuss recently discovered high-efficiency induction systems for deriving Schwann cells and neurons from MSCs. Other features of MSCs that are important for tissue repair include the self-renewing property of stem cells and their potential for differentiation. Thus, I will also discuss the stemness of MSCs and describe the discovery of a certain stem cell type among adult MSCs that can self-renew and differentiate into cells of all three germ layers. Furthermore, I will explore the prospects of using this cell population for cell therapy.
Collapse
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
35
|
Mohamad Buang ML, Seng HK, Chung LH, Saim AB, Idrus RBH. In vitro Generation of Functional Insulin-producing Cells from Lipoaspirated Human Adipose Tissue-derived Stem Cells. Arch Med Res 2012; 43:83-8. [DOI: 10.1016/j.arcmed.2012.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 01/13/2012] [Indexed: 12/24/2022]
|
36
|
In vitro generation of osteochondral differentiation of human marrow mesenchymal stem cells in novel collagen-hydroxyapatite layered scaffolds. Acta Biomater 2011; 7:3999-4006. [PMID: 21757035 DOI: 10.1016/j.actbio.2011.06.040] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 06/10/2011] [Accepted: 06/22/2011] [Indexed: 11/22/2022]
Abstract
Integrated, layered osteochondral (OC) composite materials and/or engineered OC grafts are considered as promising strategies for the treatment of OC damage. A novel biomimetic collagen-hydroxyapatite (COL-HA) OC scaffold with different integrated layers has been generated by freeze-drying. The capacity of the upper COL layer and the lower COL/HA layer to promote the growth and differentiation of human mesenchymal stem cells (hMSCs) into chondrocytes and osteoblasts respectively was evaluated. Cell viability and proliferation on COL and COL/HA scaffolds were assessed by the MTT test. The chondrogenic differentiation of hMSCs on both scaffolds was evaluated by glucosaminoglycan (GAG) quantification, alcian blue staining, type II collagen immunocytochemistry assay and real-time polymerase chain reaction in chondrogenic medium for 21 days. Osteogenic differentiation was evaluated by alkaline phosphatase activity assay, type I collagen immunocytochemistry staining, alizarin S staining and mRNA expression of osteogenic gene for 14 days in osteogenic medium. The results indicated that hMSCs on both COL and COL/HA scaffolds were viable and able to proliferate over time. The COL layer was more efficient in inducing hMSC chondrogenic differentiation than the COL/HA layer, while the COL/HA layer possessed the superiority on promoting hMSC osteogenic induction over either COL layer or pure HA. In conclusion, the layered OC composite materials can effectively promote cartilage and bone tissue generation in vitro and are potentially usable for OC tissue engineering.
Collapse
|
37
|
Liao HT, Chen CT, Chen JP. Osteogenic Differentiation and Ectopic Bone Formation of Canine Bone Marrow-Derived Mesenchymal Stem Cells in Injectable Thermo-Responsive Polymer Hydrogel. Tissue Eng Part C Methods 2011; 17:1139-49. [DOI: 10.1089/ten.tec.2011.0140] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Han-Tsung Liao
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan, Taiwan, Republic of China
- Division of Traumatic Plastic Surgery, Department of Plastic and Reconstructive Surgery, Craniofacial Research Center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-San, Taoyuan, Taiwan, Republic of China
| | - Chien-Tzung Chen
- Division of Traumatic Plastic Surgery, Department of Plastic and Reconstructive Surgery, Craniofacial Research Center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-San, Taoyuan, Taiwan, Republic of China
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
38
|
Jurgens WJFM, Lu Z, Zandieh-Doulabi B, Kuik DJ, Ritt MJPF, Helder MN. Hyperosmolarity and hypoxia induce chondrogenesis of adipose-derived stem cells in a collagen type 2 hydrogel. J Tissue Eng Regen Med 2011; 6:570-8. [PMID: 21916017 DOI: 10.1002/term.464] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 04/13/2011] [Accepted: 07/04/2011] [Indexed: 12/17/2022]
Abstract
Apart from soluble growth factors, various other biophysicochemical cues are known to promote chondrogenesis. Under physiological conditions, cartilage in the joint comprises a hyperosmotic and hypoxic environment. Therefore, in this study, we examined the inductive effects of hyperosmotic and/or hypoxic conditions on adipose stem cells (ASCs) and compared them with conventional TGFβ1-induction. After encapsulation in collagen type II hydrogels and specific induction, ASCs were assessed for viability, proliferation, morphology and chondrogenic differentiation potential. Viability was similar under all conditions, with low proliferative activity. After 4 days, hypoxia and/or hyperosmolarity did not affect round cell morphology, while cells were mainly stretched in the TGFβ1-induced group. At 21 days, the TGFß1-treated group had aggregated into a cell nodule. Hyperosmolarity mimicked this aggregation to a lesser extent, whereas cells under hypoxia stretched out after 21 days, with a combined effect in the hypoxic/hyperosmotic group. Both individual and combined hyperosmotic and/or hypoxic conditions significantly upregulated SOX5, SOX9, COMP and Link-p gene expression compared with the non-induced group, and to similar levels as the TGFβ1-induced group. GAG synthesis in both hydrogel and medium was increased under hypoxic conditions, whereas hyperosmolarity decreased GAG formation in the hydrogels, but increased GAG formation in the medium. We conclude that in a joint mimicking the three-dimensional (3D) micro-environment, a combination of hyperosmolarity and hypoxia is able to induce chondrogenesis to the same extent as TGFβ1. This might lead to an interesting alternative when considering short-term triggering in a one-step surgical procedure for the treatment of cartilaginous defects.
Collapse
Affiliation(s)
- Wouter J F M Jurgens
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Ignatius A, Schoengraf P, Kreja L, Liedert A, Recknagel S, Kandert S, Brenner RE, Schneider M, Lambris JD, Huber-Lang M. Complement C3a and C5a modulate osteoclast formation and inflammatory response of osteoblasts in synergism with IL-1β. J Cell Biochem 2011; 112:2594-605. [PMID: 21598302 PMCID: PMC3158833 DOI: 10.1002/jcb.23186] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is a tight interaction of the bone and the immune system. However, little is known about the relevance of the complement system, an important part of innate immunity and a crucial trigger for inflammation. The aim of this study was, therefore, to investigate the presence and function of complement in bone cells including osteoblasts, mesenchymal stem cells (MSC), and osteoclasts. qRT-PCR and immunostaining revealed that the central complement receptors C3aR and C5aR, complement C3 and C5, and membrane-bound regulatory proteins CD46, CD55, and CD59 were expressed in human MSC, osteoblasts, and osteoclasts. Furthermore, osteoblasts and particularly osteoclasts were able to activate complement by cleaving C5 to its active form C5a as measured by ELISA. Both C3a and C5a alone were unable to trigger the release of inflammatory cytokines interleukin (IL)-6 and IL-8 from osteoblasts. However, co-stimulation with the pro-inflammatory cytokine IL-1β significantly induced IL-6 and IL-8 expression as well as the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) indicating that complement may modulate the inflammatory response of osteoblastic cells in a pro-inflammatory environment as well as osteoblast-osteoclast interaction. While C3a and C5a did not affect osteogenic differentiation, osteoclastogenesis was significantly induced even in the absence of RANKL and macrophage-colony stimulating factor (M-CSF) suggesting that complement could directly regulate osteoclast formation. It can therefore be proposed that complement may enhance the inflammatory response of osteoblasts and increase osteoclast formation, particularly in a pro-inflammatory environment, for example, during bone healing or in inflammatory bone disorders.
Collapse
Affiliation(s)
- Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research Ulm, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Yang XF, He X, He J, Zhang LH, Su XJ, Dong ZY, Xu YJ, Li Y, Li YL. High efficient isolation and systematic identification of human adipose-derived mesenchymal stem cells. J Biomed Sci 2011; 18:59. [PMID: 21854621 PMCID: PMC3175156 DOI: 10.1186/1423-0127-18-59] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 08/19/2011] [Indexed: 01/10/2023] Open
Abstract
Background Developing efficient methods to isolate and identify human adipose-derived mesenchymal stem cells (hADSCs) remains to be one of the major challenges in tissue engineering. Methods We demonstrate here a method by isolating hADSCs from abdominal subcutaneous adipose tissue harvested during caesarian section. The hADSCs were isolated from human adipose tissue by collagenase digestion and adherence to flasks. Results The yield reached around 1 × 106 hADSCs per gram adipose tissue. The following comprehensive identification and characterization illustrated pronounced features of mesenchymal stem cells (MSCs). The fibroblast-like hADSCs exhibited typical ultrastructure details for vigorous cell activities. Karyotype mapping showed normal human chromosome. With unique immunophenotypes they were positive for CD29, CD44, CD73, CD105 and CD166, but negative for CD31, CD34, CD45 and HLA-DR. The growth curve and cell cycle analysis revealed high capability for self-renewal and proliferation. Moreover, these cells could be functionally induced into adipocytes, osteoblasts, and endothelial cells in the presence of appropriate conditioned media. Conclusion The data presented here suggest that we have developed high efficient isolation and cultivation methods with a systematic strategy for identification and characterization of hADSCs. These techniques will be able to provide safe and stable seeding cells for research and clinical application.
Collapse
Affiliation(s)
- Xu-Fang Yang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Biophysical regulation of histone acetylation in mesenchymal stem cells. Biophys J 2011; 100:1902-9. [PMID: 21504726 DOI: 10.1016/j.bpj.2011.03.008] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 02/16/2011] [Accepted: 03/09/2011] [Indexed: 11/21/2022] Open
Abstract
Histone deacetylation and acetylation are catalyzed by histone deacetylase (HDAC) and histone acetyltransferase, respectively, which play important roles in the regulation of chromatin remodeling, gene expression, and cell functions. However, whether and how biophysical cues modulate HDAC activity and histone acetylation is not well understood. Here, we tested the hypothesis that microtopographic patterning and mechanical strain on the substrate regulate nuclear shape, HDAC activity, and histone acetylation. Bone marrow mesenchymal stem cells (MSCs) were cultured on elastic membranes patterned with parallel microgrooves 10 μm wide that kept MSCs aligned along the axis of the grooves. Compared with MSCs on an unpatterned substrate, MSCs on microgrooves had elongated nuclear shape, a decrease in HDAC activity, and an increase of histone acetylation. To investigate anisotropic mechanical sensing by MSCs, cells on the elastic micropatterned membranes were subjected to static uniaxial mechanical compression or stretch in the direction parallel or perpendicular to the microgrooves. Among the four types of loads, compression or stretch perpendicular to the microgrooves caused a decrease in HDAC activity, accompanied by the increase in histone acetylation and slight changes of nuclear shape. Knocking down nuclear matrix protein lamin A/C abolished mechanical strain-induced changes in HDAC activity. These results demonstrate that micropattern and mechanical strain on the substrate can modulate nuclear shape, HDAC activity, and histone acetylation in an anisotropic manner and that nuclear matrix mediates mechanotransduction. These findings reveal a new mechanism, to our knowledge, by which extracellular biophysical signals are translated into biochemical signaling events in the nucleus, and they will have significant impact in the area of mechanobiology and mechanotransduction.
Collapse
|
42
|
Huang PI, Chen YC, Chen LH, Juan CC, Ku HH, Wang ST, Chiou SH, Chiou GY, Chi CW, Hsu CC, Lee HC, Chen LK, Kao CL. PGC-1α mediates differentiation of mesenchymal stem cells to brown adipose cells. J Atheroscler Thromb 2011; 18:966-80. [PMID: 21817823 DOI: 10.5551/jat.7401] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIM Mesenchymal stem cells (MSCs) are a multipotent cell type that can differentiate into non-hematopoietic cells, such as adipocytes. Adipocyte tissue is central to the regulation of energy balance. Two functionally different types of fat are present in mammals. White adipose tissue is the primary site for triglyceride storage, while brown adipose tissue is specialized in energy expenditure. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) controls several aspects of mitochondrial biogenesis. In this study, we hypothesized that PGC-1α plays a role in brown fat differentiation of MSCs. METHODS Immortalized human MSCs were infected with adenovirus carrying PGC-1α cDNA to create PGC-1α-expressing MSCs. RESULTS The genetic profiling of PGC-1α-expressing MSCs shows the significant increase of genes related to mitochondrial functions and lipid metabolism compared to that of MSCs. When expressed in MSCs, PGC-1α activates robust mitochondrial biogenesis and respiration. The increase of oxygen consumption and reactive oxygen species represents a cellular readout of increased activity of the respiratory chain. The expression of thermogenic markers, such as cytochrome C and complex II, was significantly increased in MSCs with treatment of adenovirus expressing PGC-1α. Moreover, PGC-1α markedly inhibited the osteogenesis of MSCs under osteogenic induction. During adipogenesis, PGC-1α-expressing MSCs showed a significant increase in brown fat markers and a decrease in white fat markers. Notably, PGC-1α knockdown inhibited adipocyte differentiation of MSCs. CONCLUSIONS In summary, our data reveal an important role of PGC-1α in promoting brown fat differentiation of MSCs, and provide a new therapeutic approach for the treatment of obesity.
Collapse
Affiliation(s)
- Pin-I Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Griffin M, Iqbal SA, Bayat A. Exploring the application of mesenchymal stem cells in bone repair and regeneration. ACTA ACUST UNITED AC 2011; 93:427-34. [PMID: 21464477 DOI: 10.1302/0301-620x.93b4.25249] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Failure of bone repair is a challenging problem in the management of fractures. There is a limited supply of autologous bone grafts for treating nonunions, with associated morbidity after harvesting. There is need for a better source of cells for repair. Mesenchymal stem cells (MSCs) hold promise for healing of bone because of their capacity to differentiate into osteoblasts and their availability from a wide variety of sources. Our review aims to evaluate the available clinical evidence and recent progress in strategies which attempt to use autologous and heterologous MSCs in clinical practice, including genetically-modified MSCs and those grown on scaffolds. We have compared various procedures for isolating and expanding a sufficient number of MSCs for use in a clinical setting. There are now a number of clinical studies which have shown that implantation of MSCs is an effective, safe and durable method for aiding the repair and regeneration of bone.
Collapse
Affiliation(s)
- M Griffin
- Plastic and Reconstructive Surgery Research, Manchester Interdisciplinary Biocentre, School of Translational Medicine, University of Manchester, Princess Street, Manchester M1 7DN, UK
| | | | | |
Collapse
|
44
|
Strategies for regeneration of the bone using porcine adult adipose-derived mesenchymal stem cells. Theriogenology 2011; 75:1381-99. [DOI: 10.1016/j.theriogenology.2010.11.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 12/17/2022]
|
45
|
Regeneration of cartilage and bone by defined subsets of mesenchymal stromal cells--potential and pitfalls. Adv Drug Deliv Rev 2011; 63:342-51. [PMID: 21184789 DOI: 10.1016/j.addr.2010.12.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 12/08/2010] [Accepted: 12/10/2010] [Indexed: 01/09/2023]
Abstract
Mesenchymal stromal cells, also referred to as mesenchymal stem cells, can be obtained from various tissues. Today the main source for isolation of mesenchymal stromal cells in mammals is the bone marrow. Mesenchymal stromal cells play an important role in tissue formation and organogenesis during embryonic development. Moreover, they provide the cellular and humoral basis for many processes of tissue regeneration and wound healing in infancy, adolescence and adulthood as well. There is increasing evidence that mesenchymal stromal cells from bone marrow and other sources including term placenta or adipose tissue are not a homogenous cell population. Only a restricted number of appropriate stem cells markers have been explored so far. But routine preparations of mesenchymal stromal cells contain phenotypically and functionally distinct subsets of stromal cells. Knowledge on the phenotypical characteristics and the functional consequences of such subsets will not only extend our understanding of stem cell biology, but might allow to develop improved regimen for regenerative medicine and wound healing and novel protocols for tissue engineering as well. In this review we will discuss novel strategies for regenerative medicine by specific selection or separation of subsets of mesenchymal stromal cells in the context of osteogenesis and bone regeneration. Mesenchymal stromal cells, which express the specific cell adhesion molecule CD146, also known as MCAM or MUC18, are prone for bone repair. Other cell surface proteins may allow the selection of chondrogenic, myogenic, adipogenic or other pre-determined subsets of mesenchymal stromal cells for improved regenerative applications as well.
Collapse
|
46
|
Laschober GT, Brunauer R, Jamnig A, Singh S, Hafen U, Fehrer C, Kloss F, Gassner R, Lepperdinger G. Age-Specific Changes of Mesenchymal Stem Cells Are Paralleled by Upregulation of CD106 Expression As a Response to an Inflammatory Environment. Rejuvenation Res 2011; 14:119-31. [DOI: 10.1089/rej.2010.1077] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Gerhard T. Laschober
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
- These authors contributed equally to this work
| | - Regina Brunauer
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
- These authors contributed equally to this work
| | - Angelika Jamnig
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | - Sarvpreet Singh
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | - Ulrich Hafen
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | - Christine Fehrer
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | - Frank Kloss
- Department for Cranio-, Maxillofacial & Oral Surgery, University Hospital Innsbruck, Innsbruck, Austria
| | - Robert Gassner
- Department for Cranio-, Maxillofacial & Oral Surgery, University Hospital Innsbruck, Innsbruck, Austria
| | - Günter Lepperdinger
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| |
Collapse
|
47
|
Tang C, Russell PJ, Martiniello-Wilks R, Rasko JEJ, Khatri A. Concise review: Nanoparticles and cellular carriers-allies in cancer imaging and cellular gene therapy? Stem Cells 2010; 28:1686-702. [PMID: 20629172 PMCID: PMC2996089 DOI: 10.1002/stem.473] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ineffective treatment and poor patient management continue to plague the arena of clinical oncology. The crucial issues include inadequate treatment efficacy due to ineffective targeting of cancer deposits, systemic toxicities, suboptimal cancer detection and disease monitoring. This has led to the quest for clinically relevant, innovative multifaceted solutions such as development of targeted and traceable therapies. Mesenchymal stem cells (MSCs) have the intrinsic ability to "home" to growing tumors and are hypoimmunogenic. Therefore, these can be used as (a) "Trojan Horses" to deliver gene therapy directly into the tumors and (b) carriers of nanoparticles to allow cell tracking and simultaneous cancer detection. The camouflage of MSC carriers can potentially tackle the issues of safety, vector, and/or transgene immunogenicity as well as nanoparticle clearance and toxicity. The versatility of the nanotechnology platform could allow cellular tracking using single or multimodal imaging modalities. Toward that end, noninvasive magnetic resonance imaging (MRI) is fast becoming a clinical favorite, though there is scope for improvement in its accuracy and sensitivity. In that, use of superparamagnetic iron-oxide nanoparticles (SPION) as MRI contrast enhancers may be the best option for tracking therapeutic MSC. The prospects and consequences of synergistic approaches using MSC carriers, gene therapy, and SPION in developing cancer diagnostics and therapeutics are discussed.
Collapse
Affiliation(s)
- Catherine Tang
- Oncology Research Centre, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
48
|
Jezierski A, Gruslin A, Tremblay R, Ly D, Smith C, Turksen K, Sikorska M, Bani-Yaghoub M. Probing stemness and neural commitment in human amniotic fluid cells. Stem Cell Rev Rep 2010; 6:199-214. [PMID: 20221716 DOI: 10.1007/s12015-010-9116-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently, human amniotic fluid (AF) cells have attracted a great deal of attention as an alternative cell source for transplantation and tissue engineering. AF contains a variety of cell types derived from fetal tissues, of which a small percentage is believed to represent stem cell sub-population(s). In contrast to human embryonic stem (ES) cells, AF cells are not subject to extensive legal or ethical considerations; nor are they limited by lineage commitment characteristic of adult stem cells. However, to become therapeutically valuable, better protocols for the isolation of AF stem cell sub-populations need to be developed. This study was designed to examine the molecular components involved in self-renewal, neural commitment and differentiation of AF cells obtained at different gestational ages. Our results showed that, although morphologically heterogeneous, AF cells derived from early gestational periods ubiquitously expressed KERATIN 8 (K8), suggesting that the majority of these cells may have an epithelial origin. In addition, AF cells expressed various components of NOTCH signaling (ligands, receptors and target genes), a pathway involved in stem cell maintenance, determination and differentiation. A sub-population of K8 positive cells (<10%) co-expressed NESTIN, a marker detected in the neuroepithelium, neural stem cells and neural progenitors. Throughout the gestational periods, a much smaller AF cell sub-population (<1%) expressed pluripotency markers, OCT4a, NANOG and SOX2, from which SOX2 positive AF cells could be isolated through single cell cloning. The SOX2 expressing AF clones showed the capacity to give rise to a neuron-like phenotype in culture, expressing neuronal markers such as MAP2, NFL and NSE. Taken together, our findings demonstrated the presence of fetal cells with stem cell characteristics in the amniotic fluid, highlighting the need for further research on their biology and clinical applications.
Collapse
Affiliation(s)
- Anna Jezierski
- Neurogenesis and Brain Repair, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Road, Ottawa, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Klopp AH, Lacerda L, Gupta A, Debeb BG, Solley T, Li L, Spaeth E, Xu W, Zhang X, Lewis MT, Reuben JM, Krishnamurthy S, Ferrari M, Gaspar R, Buchholz TA, Cristofanilli M, Marini F, Andreeff M, Woodward WA. Mesenchymal stem cells promote mammosphere formation and decrease E-cadherin in normal and malignant breast cells. PLoS One 2010; 5:e12180. [PMID: 20808935 PMCID: PMC2922340 DOI: 10.1371/journal.pone.0012180] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 07/26/2010] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Normal and malignant breast tissue contains a rare population of multi-potent cells with the capacity to self-renew, referred to as stem cells, or tumor initiating cells (TIC). These cells can be enriched by growth as "mammospheres" in three-dimensional cultures. OBJECTIVE We tested the hypothesis that human bone-marrow derived mesenchymal stem cells (MSC), which are known to support tumor growth and metastasis, increase mammosphere formation. RESULTS We found that MSC increased human mammary epithelial cell (HMEC) mammosphere formation in a dose-dependent manner. A similar increase in sphere formation was seen in human inflammatory (SUM149) and non-inflammatory breast cancer cell lines (MCF-7) but not in primary inflammatory breast cancer cells (MDA-IBC-3). We determined that increased mammosphere formation can be mediated by secreted factors as MSC conditioned media from MSC spheroids significantly increased HMEC, MCF-7 and SUM149 mammosphere formation by 6.4 to 21-fold. Mammospheres grown in MSC conditioned media had lower levels of the cell adhesion protein, E-cadherin, and increased expression of N-cadherin in SUM149 and HMEC cells, characteristic of a pro-invasive mesenchymal phenotype. Co-injection with MSC in vivo resulted in a reduced latency time to develop detectable MCF-7 and MDA-IBC-3 tumors and increased the growth of MDA-IBC-3 tumors. Furthermore, E-cadherin expression was decreased in MDA-IBC-3 xenografts with co-injection of MSC. CONCLUSIONS MSC increase the efficiency of primary mammosphere formation in normal and malignant breast cells and decrease E-cadherin expression, a biologic event associated with breast cancer progression and resistance to therapy.
Collapse
Affiliation(s)
- Ann H. Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lara Lacerda
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of NanoMedicine and Biomedical Engineering, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Anshul Gupta
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bisrat G. Debeb
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Travis Solley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Li Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Erika Spaeth
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Wei Xu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiaomei Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Mauro Ferrari
- Department of NanoMedicine and Biomedical Engineering, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Rogério Gaspar
- iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Thomas A. Buchholz
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Massimo Cristofanilli
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Frank Marini
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Andreeff
- Department of Stem Cell Transplant and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
50
|
You MH, Kwak MK, Kim DH, Kim K, Levchenko A, Kim DY, Suh KY. Synergistically enhanced osteogenic differentiation of human mesenchymal stem cells by culture on nanostructured surfaces with induction media. Biomacromolecules 2010; 11:1856-62. [PMID: 20568737 PMCID: PMC2921863 DOI: 10.1021/bm100374n] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We have examined the effects of surface nanotopography on in vitro osteogenesis of human mesenchymal stem cells (hMSCs). UV-assisted capillary force lithography was employed to fabricate a scalable (4x5 cm), well-defined nanostructured substrate of a UV curable polyurethane polymer with dots (150, 400, 600 nm diameter) and lines (150, 400, 600 nm width). The influence of osteogenic differentiation of hMSCs was characterized at day 8 by alkaline phosphatase (ALP) assay, RT-PCR, and real-time PCR analysis. We found that hMSCs cultured on the nanostructured surfaces in osteogenic induction media showed significantly higher ALP activity compared to unpatterned PUA surface (control group). In particular, the hMSCs on the 400 nm dot pattern showed the highest level of ALP activity. Further investigation with real-time quantitative RT-PCR analysis demonstrated significantly higher expression of core binding factor 1 (Cbfa1), osteopontin (OP), and osteocalcin (OC) levels in hMSCs cultured on the 400 nm dot pattern in osteogenic induction media. These findings suggest that surface nanotopography can enhance osteogenic differentiation synergistically with biochemical induction substance.
Collapse
Affiliation(s)
- Mi-Hyeon You
- Department of Veterinary Pathology, Seoul National University, Seoul 151-742, Korea
| | - Moon Kyu Kwak
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Korea
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Keesung Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Korea
| | - Andre Levchenko
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore MD 21218, USA
| | - Dae-Yong Kim
- Department of Veterinary Pathology, Seoul National University, Seoul 151-742, Korea
| | - Kahp-Yang Suh
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Korea
- World Class University (WCU) Program on Multiscale Mechanical Design, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|