51
|
Abstract
Temporomandibular Disorders (TMD) represent a heterogeneous group of musculoskeletal and neuromuscular conditions involving the temporomandibular joint (TMJ), masticatory muscles and/or associated structures. They are a major cause of non-dental orofacial pain. As a group, they are often multi-factorial in nature and have no common etiology or biological explanations. TMD can be broadly divided into masticatory muscle and TMJ disorders. TMJ disorders are characterized by intra-articular positional and/or structural abnormalities. The most common type of TMJ disorders involves displacement of the TMJ articular disc that precedes progressive degenerative changes of the joint leading to osteoarthritis (OA). In the past decade, progress made in the development of stem cell-based therapies and tissue engineering have provided alternative methods to attenuate the disease symptoms and even replace the diseased tissue in the treatment of TMJ disorders. Resident mesenchymal stem cells (MSCs) have been isolated from the synovia of TMJ, suggesting an important role in the repair and regeneration of TMJ. The seminal discovery of pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have provided promising cell sources for drug discovery, transplantation as well as for tissue engineering of TMJ condylar cartilage and disc. This review discusses the most recent advances in development of stem cell-based treatments for TMJ disorders through innovative approaches of cell-based therapeutics, tissue engineering and drug discovery.
Collapse
|
52
|
Khoo BY, Nadarajan K, Shim SY, Miswan N, Zang CB, Possinger K, Elstner E. Pretreatment of BMSCs with TZD solution decreases the proliferation rate of MCF‑7 cells by reducing FGF4 protein expression. Mol Med Rep 2016; 13:3406-14. [PMID: 26934829 PMCID: PMC4805074 DOI: 10.3892/mmr.2016.4959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 02/04/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of bone marrow‑derived mesenchymal stem cells (BMSCs) that had been pretreated with pioglitazone and/or rosiglitazone on the growth and proliferation rate of MCF‑7 cells. The adhesive interaction between the BMSCs and the MCF‑7 cancer cells revealed that the pretreatment of BMSCs with a combination of two types of thiazolidinedione drug reduced the growth and proliferation rate of the MCF‑7 cells. The proliferation rate of the MCF‑7 cells could also be reduced by the non‑adhesive interaction of the cancer cells with BMSCs pretreated with pioglitazone and/or rosiglitazone. The growth and proliferation rate reduction effects on the MCF‑7 cells may be attributed to the reduction in the protein level of fibroblast growth factor 4 (FGF4) in the conditioned medium of the pretreated BMSCs. The evidence that the low protein level of FGF4 in the conditioned medium of the pretreated BMSCs perturbed the proliferation rate of the MCF‑7 cells by reducing the levels of Ki‑67 and proliferating cell nuclear antigen transcripts in the cancer cells was also demonstrated in the present study using a FGF4‑neutralizing antibody. All the above findings demonstrate that future studies on the correlation between FGF4 and pretreated BMSCs would be beneficial.
Collapse
Affiliation(s)
- Boon-Yin Khoo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Kalpanah Nadarajan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Siang-Yian Shim
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Noorizan Miswan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Chuan-Bing Zang
- Division of Oncology and Haematology, Charité Campus Mitte, Humboldt University of Berlin, D‑10099 Berlin, Germany
| | - Kurt Possinger
- Division of Oncology and Haematology, Charité Campus Mitte, Humboldt University of Berlin, D‑10099 Berlin, Germany
| | - Elena Elstner
- Division of Oncology and Haematology, Charité Campus Mitte, Humboldt University of Berlin, D‑10099 Berlin, Germany
| |
Collapse
|
53
|
Manufacturing of Human Umbilical Cord Mesenchymal Stromal Cells on Microcarriers in a Dynamic System for Clinical Use. Stem Cells Int 2016; 2016:4834616. [PMID: 26977155 PMCID: PMC4761675 DOI: 10.1155/2016/4834616] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 12/15/2022] Open
Abstract
The great properties of human mesenchymal stromal cells (hMSCs) make these cells an important tool in regenerative medicine. Because of the limitations of hMSCs derived from the bone marrow during isolation and expansion, hMSCs derived from the umbilical cord stroma are a great alternative to overcome these issues. For a large expansion of these cells, we performed a process transfer from static culture to a dynamic system. For this reason, a microcarrier selection out of five microcarrier types was made to achieve a suitable growth surface for the cells. The growth characteristics and metabolite consumption and production were used to compare the cells growth in 12-well plate and spinner flask. The goal to determine relevant process parameters to transfer the expansion process into a stirred tank bioreactor was achieved.
Collapse
|
54
|
Standardizing Umbilical Cord Mesenchymal Stromal Cells for Translation to Clinical Use: Selection of GMP-Compliant Medium and a Simplified Isolation Method. Stem Cells Int 2016; 2016:6810980. [PMID: 26966439 PMCID: PMC4757747 DOI: 10.1155/2016/6810980] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/29/2015] [Indexed: 12/15/2022] Open
Abstract
Umbilical cord derived mesenchymal stromal cells (UC-MSCs) are a focus for clinical translation but standardized methods for isolation and expansion are lacking. Previously we published isolation and expansion methods for UC-MSCs which presented challenges when considering good manufacturing practices (GMP) for clinical translation. Here, a new and more standardized method for isolation and expansion of UC-MSCs is described. The new method eliminates dissection of blood vessels and uses a closed-vessel dissociation following enzymatic digestion which reduces contamination risk and manipulation time. The new method produced >10 times more cells per cm of UC than our previous method. When biographical variables were compared, more UC-MSCs per gram were isolated after vaginal birth compared to Caesarian-section births, an unexpected result. UC-MSCs were expanded in medium enriched with 2%, 5%, or 10% pooled human platelet lysate (HPL) eliminating the xenogeneic serum components. When the HPL concentrations were compared, media supplemented with 10% HPL had the highest growth rate, smallest cells, and the most viable cells at passage. UC-MSCs grown in 10% HPL had surface marker expression typical of MSCs, high colony forming efficiency, and could undergo trilineage differentiation. The new protocol standardizes manufacturing of UC-MSCs and enables clinical translation.
Collapse
|
55
|
Karaöz E, İnci Ç. Umbilical Cord Tissue and Wharton’s Jelly Mesenchymal Stem Cells Properties and Therapeutic Potentials. PERINATAL TISSUE-DERIVED STEM CELLS 2016. [DOI: 10.1007/978-3-319-46410-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
56
|
|
57
|
Sridharan B, Lin SM, Hwu AT, Laflin AD, Detamore MS. Stem Cells in Aggregate Form to Enhance Chondrogenesis in Hydrogels. PLoS One 2015; 10:e0141479. [PMID: 26719986 PMCID: PMC4697858 DOI: 10.1371/journal.pone.0141479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
There are a variety of exciting hydrogel technologies being explored for cartilage regenerative medicine. Our overall goal is to explore whether using stem cells in an aggregate form may be advantageous in these applications. 3D stem cell aggregates hold great promise as they may recapitulate the in vivo skeletal tissue condensation, a property that is not typically observed in 2D culture. We considered two different stem cell sources, human umbilical cord Wharton’s jelly cells (hWJCs, currently being used in clinical trials) and rat bone marrow-derived mesenchymal stem cells (rBMSCs). The objective of the current study was to compare the influence of cell phenotype, aggregate size, and aggregate number on chondrogenic differentiation in a generic hydrogel (agarose) platform. Despite being differing cell sources, both rBMSC and hWJC aggregates were consistent in outperforming cell suspension control groups in biosynthesis and chondrogenesis. Higher cell density impacted biosynthesis favorably, and the number of aggregates positively influenced chondrogenesis. Therefore, we recommend that investigators employing hydrogels consider using cells in an aggregate form for enhanced chondrogenic performance.
Collapse
Affiliation(s)
- BanuPriya Sridharan
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
| | - Staphany M. Lin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Alexander T. Hwu
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Amy D. Laflin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Michael S. Detamore
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
58
|
Reppel L, Schiavi J, Charif N, Leger L, Yu H, Pinzano A, Henrionnet C, Stoltz JF, Bensoussan D, Huselstein C. Chondrogenic induction of mesenchymal stromal/stem cells from Wharton's jelly embedded in alginate hydrogel and without added growth factor: an alternative stem cell source for cartilage tissue engineering. Stem Cell Res Ther 2015; 6:260. [PMID: 26718750 PMCID: PMC4697319 DOI: 10.1186/s13287-015-0263-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022] Open
Abstract
Background Due to their intrinsic properties, stem cells are promising tools for new developments in tissue engineering and particularly for cartilage tissue regeneration. Although mesenchymal stromal/stem cells from bone marrow (BM-MSC) have long been the most used stem cell source in cartilage tissue engineering, they have certain limits. Thanks to their properties such as low immunogenicity and particularly chondrogenic differentiation potential, mesenchymal stromal/stem cells from Wharton’s jelly (WJ-MSC) promise to be an interesting source of MSC for cartilage tissue engineering. Methods In this study, we propose to evaluate chondrogenic potential of WJ-MSC embedded in alginate/hyaluronic acid hydrogel over 28 days. Hydrogels were constructed by the original spraying method. Our main objective was to evaluate chondrogenic differentiation of WJ-MSC on three-dimensional scaffolds, without adding growth factors, at transcript and protein levels. We compared the results to those obtained from standard BM-MSC. Results After 3 days of culture, WJ-MSC seemed to be adapted to their new three-dimensional environment without any detectable damage. From day 14 and up to 28 days, the proportion of WJ-MSC CD73+, CD90+, CD105+ and CD166+ decreased significantly compared to monolayer marker expression. Moreover, WJ-MSC and BM-MSC showed different phenotype profiles. After 28 days of scaffold culture, our results showed strong upregulation of cartilage-specific transcript expression. WJ-MSC exhibited greater type II collagen synthesis than BM-MSC at both transcript and protein levels. Furthermore, our work highlighted a relevant result showing that WJ-MSC expressed Runx2 and type X collagen at lower levels than BM-MSC. Conclusions Once seeded in the hydrogel scaffold, WJ-MSC and BM-MSC have different profiles of chondrogenic differentiation at both the phenotypic level and matrix synthesis. After 4 weeks, WJ-MSC, embedded in a three-dimensional environment, were able to adapt to their environment and express specific cartilage-related genes and matrix proteins. Today, WJ-MSC represent a real alternative source of stem cells for cartilage tissue engineering.
Collapse
Affiliation(s)
- Loïc Reppel
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,CHU de Nancy, Unité de Thérapie Cellulaire et Tissulaire, 54500, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Jessica Schiavi
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Naceur Charif
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Léonore Leger
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Hao Yu
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Astrid Pinzano
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Christel Henrionnet
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Jean-François Stoltz
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,CHU de Nancy, Unité de Thérapie Cellulaire et Tissulaire, 54500, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Danièle Bensoussan
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,CHU de Nancy, Unité de Thérapie Cellulaire et Tissulaire, 54500, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| | - Céline Huselstein
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54505, Vandœuvre-lès-Nancy, France. .,Université de Lorraine, 54000, Nancy, France. .,Fédération de Recherche 3209, Bioingénierie Moléculaire Cellulaire et Thérapeutique, 54500, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
59
|
Park YB, Song M, Lee CH, Kim JA, Ha CW. Cartilage repair by human umbilical cord blood-derived mesenchymal stem cells with different hydrogels in a rat model. J Orthop Res 2015; 33:1580-6. [PMID: 26019012 DOI: 10.1002/jor.22950] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/25/2015] [Indexed: 02/04/2023]
Abstract
This study was carried out to assess the feasibility of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) in articular cartilage repair and to further determine a suitable delivering hydrogel in a rat model. Critical sized full thickness cartilage defects were created. The hUCB-MSCs and three different hydrogel composites (hydrogel A; 4% hyaluronic acid/30% pluronic (1:1, v/v), hydrogel B; 4% hyaluronic acid, and hydrogel C; 4% hyaluronic acid/30% pluronic/chitosan (1:1:2, v/v)) were implanted into the experimental knee (right knee) and hydrogels without hUCB-MSCs were implanted into the control knee (left knee). Defects were evaluated after 8 weeks. The hUCB-MSCs with hydrogels composites resulted in a better repair as seen by gross and histological evaluation compared with hydrogels without hUCB-MSCs. Among the three different hydrogels, the 4% hyaluronic acid hydrogel composite (hydrogel B) showed the best result in cartilage repair as seen by the histological evaluation compared with the other hydrogel composites (hydrogel A and C). The results of this study suggest that hUCB-MSCs may be a promising cell source in combination with 4% hyaluronic acid hydrogels in the in vivo repair of cartilage defects.
Collapse
Affiliation(s)
- Yong-Beom Park
- Department of Orthopedic Surgery, Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Minjung Song
- Department of Orthopedic Surgery, Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Choong-Hee Lee
- Department of Orthopedic Surgery, Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin-A Kim
- Department of Orthopedic Surgery, Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Chul-Won Ha
- Department of Orthopedic Surgery, Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
60
|
Pereira RC, Costa-Pinto AR, Frias AM, Neves NM, Azevedo HS, Reis RL. In vitro chondrogenic commitment of human Wharton's jelly stem cells by co-culture with human articular chondrocytes. J Tissue Eng Regen Med 2015; 11:1876-1887. [PMID: 27035732 DOI: 10.1002/term.2085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/11/2015] [Indexed: 11/09/2022]
Abstract
Wharton's jelly stem cells (WJSCs) are a potential source of transplantable stem cells in cartilage-regenerative strategies, due to their highly proliferative and multilineage differentiation capacity. We hypothesized that a non-direct co-culture system with human articular chondrocytes (hACs) could enhance the potential chondrogenic phenotype of hWJSCs during the expansion phase compared to those expanded in monoculture conditions. Primary hWJSCs were cultured in the bottom of a multiwell plate separated by a porous transwell membrane insert seeded with hACs. No statistically significant differences in hWJSCs duplication number were observed under either of the culture conditions during the expansion phase. hWJSCs under co-culture conditions show upregulations of collagen type I and II, COMP, TGFβ1 and aggrecan, as well as of the main cartilage transcription factor, SOX9, when compared to those cultured in the absence of chondrocytes. Chondrogenic differentiation of hWJSCs, previously expanded in co-culture and monoculture conditions, was evaluated for each cellular passage using the micromass culture model. Cells expanded in co-culture showed higher accumulation of glycosaminoglycans (GAGs) compared to cells in monoculture, and immunohistochemistry for localization of collagen type I revealed a strong detection signal when hWJSCs were expanded under monoculture conditions. In contrast, type II collagen was detected when cells were expanded under co-culture conditions, where numerous round-shaped cell clusters were observed. Using a micromass differentiation model, hWJSCs, previously exposed to soluble factors secreted by hACs, were able to express higher levels of chondrogenic genes with deposition of cartilage extracellular matrix components, suggesting their use as an alternative cell source for treating degenerated cartilage. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- R C Pereira
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| | - A R Costa-Pinto
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| | - A M Frias
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| | - N M Neves
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| | - H S Azevedo
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| | - R L Reis
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Guimarães, Portugal.,ICVS/3Bs PT Government-associated Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
61
|
Klontzas ME, Kenanidis EI, Heliotis M, Tsiridis E, Mantalaris A. Bone and cartilage regeneration with the use of umbilical cord mesenchymal stem cells. Expert Opin Biol Ther 2015; 15:1541-52. [PMID: 26176327 DOI: 10.1517/14712598.2015.1068755] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION The production of functional alternatives to bone autografts and the development new treatment strategies for cartilage defects are great challenges that could be addressed by the field of tissue engineering. Umbilical cord mesenchymal stem cells (MSCs) can be used to produce cost-effective, atraumatic and possibly autologous bone and cartilage grafts. AREAS COVERED MSCs can be isolated from umbilical cord Wharton's jelly, perivascular tissue and blood using various techniques. Those cells have been characterized and phenotypic similarities with bone marrow-derived MSCs (BM-MSCs) and embryonic stem cells have been found. Findings on their differentiation into the osteogenic and chondrogenic lineage differ between studies and are not as consistent as for BM-MSCs. EXPERT OPINION MSCs from umbilical cords have to be more extensively studied and the mechanisms underlying their differentiation have to be clarified. To date, they seem to be an attractive alternative to BM-MSCs. However, further research with suitable scaffolds and growth factors as well as with novel scaffold fabrication and culture technology should be conducted before they are introduced to clinical practice and replace BM-MSCs.
Collapse
Affiliation(s)
- Michail E Klontzas
- a 1 Imperial College London, Department of Chemical Engineering and Chemical Technology , South Kensington Campus, London, UK
| | - Eustathios I Kenanidis
- b 2 Aristotle University Medical School, Academic Orthopaedic Unit , University Campus 54 124, Thessaloniki, Greece.,c 3 Aristotle University Medical School, "PapaGeorgiou" General Hospital, Academic Orthopaedic Unit , Thessaloniki, Greece
| | | | - Eleftherios Tsiridis
- b 2 Aristotle University Medical School, Academic Orthopaedic Unit , University Campus 54 124, Thessaloniki, Greece.,e 5 Imperial College London, Department of Surgery and Cancer, Division of Surgery , B-block, Hammersmith, Du-Cane Road, London, UK
| | - Athanasios Mantalaris
- f 6 Imperial College London, Department of Chemical Engineering , South Kensington Campus, London, UK
| |
Collapse
|
62
|
A 3D Porous Gelatin-Alginate-Based-IPN Acts as an Efficient Promoter of Chondrogenesis from Human Adipose-Derived Stem Cells. Stem Cells Int 2015; 2015:252909. [PMID: 26106422 PMCID: PMC4461772 DOI: 10.1155/2015/252909] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/18/2015] [Indexed: 12/30/2022] Open
Abstract
Cartilage has limited regeneration potential. Thus, there is an imperative need to develop new strategies for cartilage tissue engineering (CTE) amenable for clinical use. Recent CTE approaches rely on optimal cell-scaffold interactions, which require a great deal of optimization. In this study we attempt to build a novel gelatin- (G-) alginate- (A-) polyacrylamide (PAA) 3D interpenetrating network (IPN) with superior performance in promoting chondrogenesis from human adipose-derived stem cells (hADSCs). We show that our G-A-PAA scaffold is capable of supporting hADSCs proliferation and survival, with no apparent cytotoxic effect. Moreover, we find that after exposure to prochondrogenic conditions a key transcription factor known to induce chondrogenesis, namely, Sox9, is highly expressed in our hADSCs/G-A-PAA bioconstruct, along with cartilage specific markers such as collagen type II, CEP68, and COMP extracellular matrix (ECM) components. These data suggest that our G-A-PAA structural properties and formulation might enable hADSCs conversion towards functional chondrocytes. We conclude that our novel G-A-PAA biomatrix is a good candidate for prospective in vivo CTE applications.
Collapse
|
63
|
McGuirk JP, Smith JR, Divine CL, Zuniga M, Weiss ML. Wharton's Jelly-Derived Mesenchymal Stromal Cells as a Promising Cellular Therapeutic Strategy for the Management of Graft-versus-Host Disease. Pharmaceuticals (Basel) 2015; 8:196-220. [PMID: 25894816 PMCID: PMC4491656 DOI: 10.3390/ph8020196] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/13/2015] [Accepted: 04/08/2015] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT), a treatment option in hematologic malignancies and bone marrow failure syndromes, is frequently complicated by Graft-versus-host disease (GVHD). The primary treatment for GVHD involves immune suppression by glucocorticoids. However, patients are often refractory to the steroid therapy, and this results in a poor prognosis. Therefore alternative therapies are needed to treat GVHD. Here, we review data supporting the clinical investigation of a novel cellular therapy using Wharton’s jelly (WJ)-derived mesenchymal stromal cells (MSCs) as a potentially safe and effective therapeutic strategy in the management of GVHD. Adult-derived sources of MSCs have demonstrated signals of efficacy in the management of GVHD. However, there are limitations, including: limited proliferation capacity; heterogeneity of cell sources; lengthy expansion time to clinical dose; expansion failure in vitro; and a painful, invasive, isolation procedure for the donor. Therefore, alternative MSC sources for cellular therapy are sought. The reviewed data suggests MSCs derived from WJ may be a safe and effective cellular therapy for GVHD. Laboratories investigated and defined the immune properties of WJ-MSCs for potential use in cellular therapy. These cells represent a more uniform cell population than bone marrow-derived MSCs, displaying robust immunosuppressive properties and lacking significant immunogenicity. They can be collected safely and painlessly from individuals at birth, rapidly expanded and stored cryogenically for later clinical use. Additionally, data we reviewed suggested licensing MSCs (activating MSCs by exposure to cytokines) to enhance effectiveness in treating GVHD. Therefore, WJCs should be tested as a second generation, relatively homogeneous allogeneic cell therapy for the treatment of GVHD.
Collapse
Affiliation(s)
- Joseph P McGuirk
- Blood and Marrow Transplant Program, The University of Kansas Medical Center, 2330 Shawnee Mission Pkwy., Suite 210 Mailstop 5003, Westwood, KS 66205, USA.
| | - J Robert Smith
- Department of Anatomy and Physiology, Kansas State University, 1600 Denison Ave., Coles Hall 228, Manhattan, KS 66506-5802, USA.
| | - Clint L Divine
- Blood and Marrow Transplant Program, The University of Kansas Medical Center, 2330 Shawnee Mission Pkwy., Suite 210 Mailstop 5003, Westwood, KS 66205, USA
| | - Micheal Zuniga
- Department of Anatomy and Physiology, Kansas State University, 1600 Denison Ave., Coles Hall 228, Manhattan, KS 66506-5802, USA.
| | - Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, 1600 Denison Ave., Coles Hall 228, Manhattan, KS 66506-5802, USA.
| |
Collapse
|
64
|
Cipriani P, Ruscitti P, Di Benedetto P, Carubbi F, Liakouli V, Berardicurti O, Ciccia F, Triolo G, Giacomelli R. Mesenchymal stromal cells and rheumatic diseases: new tools from pathogenesis to regenerative therapies. Cytotherapy 2015; 17:832-49. [PMID: 25680301 DOI: 10.1016/j.jcyt.2014.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 01/08/2023]
Abstract
In recent years, mesenchymal stromal cells (MSCs) have been largely investigated and tested as a new therapeutic tool for several clinical applications, including the treatment of different rheumatic diseases. MSCs are responsible for the normal turnover and maintenance of adult mesenchymal tissues as the result of their multipotent differentiation abilities and their secretion of a variety of cytokines and growth factors. Although initially derived from bone marrow, MSCs are present in many different tissues such as many peri-articular tissues. MSCs may exert immune-modulatory properties, modulating different immune cells in both in vitro and in vivo models, and they are considered immune-privileged cells. At present, these capacities are considered the most intriguing aspect of their biology, introducing the possibility that these cells may be used as effective therapy in autoimmune diseases. Therefore, stem cell therapies may represent an innovative approach for the treatment of rheumatic diseases, especially for the forms that are not responsive to standard treatments or alternatively still lacking a definite therapy. At present, although the data from scientific literature appear to suggest that such treatments might be more effective whether administered as soon as possible, the use of MSCs in clinical practice is likely to be restricted to patients with a long history of a severe refractory disease. Further results from larger clinical trials are needed to corroborate preclinical findings and human non-controlled studies, and advancement in the knowledge of MSCs might provide information about the therapeutic role of these cells in the treatment of many rheumatic diseases.
Collapse
Affiliation(s)
- Paola Cipriani
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy.
| | - Piero Ruscitti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Paola Di Benedetto
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Carubbi
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Vasiliki Liakouli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ciccia
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Giovanni Triolo
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
65
|
Si JW, Wang XD, Shen SGF. Perinatal stem cells: A promising cell resource for tissue engineering of craniofacial bone. World J Stem Cells 2015; 7:149-159. [PMID: 25621114 PMCID: PMC4300925 DOI: 10.4252/wjsc.v7.i1.149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/28/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023] Open
Abstract
In facing the mounting clinical challenge and suboptimal techniques of craniofacial bone defects resulting from various conditions, such as congenital malformations, osteomyelitis, trauma and tumor resection, the ongoing research of regenerative medicine using stem cells and concurrent advancement in biotechnology have shifted the focus from surgical reconstruction to a novel stem cell-based tissue engineering strategy for customized and functional craniofacial bone regeneration. Given the unique ontogenetical and cell biological properties of perinatal stem cells, emerging evidence has suggested these extraembryonic tissue-derived stem cells to be a promising cell source for extensive use in regenerative medicine and tissue engineering. In this review, we summarize the current achievements and obstacles in stem cell-based craniofacial bone regeneration and subsequently we address the characteristics of various types of perinatal stem cells and their novel application in tissue engineering of craniofacial bone. We propose the promising feasibility and scope of perinatal stem cell-based craniofacial bone tissue engineering for future clinical application.
Collapse
|
66
|
The effect of extended passaging on the phenotype and osteogenic potential of human umbilical cord mesenchymal stem cells. Mol Cell Biochem 2015; 401:155-64. [PMID: 25555467 DOI: 10.1007/s11010-014-2303-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/10/2014] [Indexed: 12/21/2022]
Abstract
Retaining biological characteristics in the extended passaging is crucial for human umbilical cord mesenchymal stem cells (hUCMSCs) in tissue engineering. We aimed to assess morphology, viability, MSC marker expression, and osteogenic activity of hUCSMCs after extended passaging. Passages 4 (P4) and 16 (P16) hUCMSCs displayed similar morphology and viability. The flow cytometry results showed that CD73, CD90, and CD105 were highly expressed at P1-P16. CD166 expression decreased progressively from 90 % at P2 to 61.5 % at P5 (p < 0.05), followed by stable expression through P16. Results from calcium deposition alkaline phosphatase activity and RT-PCR assay showed that both P4 and P16 hUCMSCs differentiated down an osteogenic lineage, with no significant difference in osteogenic capacity (p < 0.05). High-passage UMCSCs maintained stable expression of MSC CD markers as well as stable osteogenic activity. hUCMSCs may thus be suitable for tissue engineering and regenerative medicine applications.
Collapse
|
67
|
Wen Y, Gu W, Cui J, Yu M, Zhang Y, Tang C, Yang P, Xu X. Platelet-rich plasma enhanced umbilical cord mesenchymal stem cells-based bone tissue regeneration. Arch Oral Biol 2014; 59:1146-54. [DOI: 10.1016/j.archoralbio.2014.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 04/30/2014] [Accepted: 07/03/2014] [Indexed: 01/05/2023]
|
68
|
Bhardwaj N, Devi D, Mandal BB. Tissue-engineered cartilage: the crossroads of biomaterials, cells and stimulating factors. Macromol Biosci 2014; 15:153-82. [PMID: 25283763 DOI: 10.1002/mabi.201400335] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/25/2014] [Indexed: 02/06/2023]
Abstract
Damage to cartilage represents one of the most challenging tasks of musculoskeletal therapeutics due to its limited propensity for healing and regenerative capabilities. Lack of current treatments to restore cartilage tissue function has prompted research in this rapidly emerging field of tissue regeneration of functional cartilage tissue substitutes. The development of cartilaginous tissue largely depends on the combination of appropriate biomaterials, cell source, and stimulating factors. Over the years, various biomaterials have been utilized for cartilage repair, but outcomes are far from achieving native cartilage architecture and function. This highlights the need for exploration of suitable biomaterials and stimulating factors for cartilage regeneration. With these perspectives, we aim to present an overview of cartilage tissue engineering with recent progress, development, and major steps taken toward the generation of functional cartilage tissue. In this review, we have discussed the advances and problems in tissue engineering of cartilage with strong emphasis on the utilization of natural polymeric biomaterials, various cell sources, and stimulating factors such as biophysical stimuli, mechanical stimuli, dynamic culture, and growth factors used so far in cartilage regeneration. Finally, we have focused on clinical trials, recent innovations, and future prospects related to cartilage engineering.
Collapse
Affiliation(s)
- Nandana Bhardwaj
- Seri-Biotechnology Unit, Life Science Division, Institute of Advanced Study in Science and Technology, Guwahati, 781035, India
| | | | | |
Collapse
|
69
|
Lei SH, Guo L, Yue HY, Zhao DC, Zhang CJ, Du WJ, Huang LZ, Wang J, Dang YX, Liu JS, Hao JL, Wang YL. Marrow stromal stem cell autologous transplantation in denervated fracture healing: an experimental study in rats. Orthop Surg 2014; 5:280-8. [PMID: 24254452 DOI: 10.1111/os.12071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 07/30/2013] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To investigate the influence of bone marrow stromal stem cell (BMSCs) transplantation on healing of fractures combined with central nerve injuries in rats. METHODS Forty-eight healthy adult SD male rats were randomly divided into the following three groups (16 rats in each group): group A, simple (left) tibial fracture; group B, tibial fracture combined with T10 spinal cord transection (SCT); group C, tibial fracture combined with T10 SCT and BMSCs transplantation. The tibial fractures were stabilized with modular intramedullary nails and all operated hind limbs were further immobilized in plaster casts to prevent unequal load bearing. BMSCs were labeled with bromodeoxyuridine and implanted into the fractures of C group rats 2 days after creation of the model. The animals in B and C groups were evaluated by postoperative Tarlov scores. The fractured tibiae were evaluated separately radiographically (X-ray and CT) and immunohistochemically 1, 2, 3 and 4 weeks after injury to assess fracture healing. In addition, the wet weights of the left tibias were measured. RESULTS All Tarlov score of the B and C group animals reached the requirements of the experiment. One, 2 and 3 weeks after surgery, the tibial callus widths in B and C group animals were significantly greater than those of group A rats (P < 0.05). At 4 weeks the tibial callus width in group C animals had decreased, but still differed significantly from that in group A rats (P < 0.05). One, 2, 3 and 4 weeks after surgery, the wet weights of B and C group tibias were significantly greater than those of group A (P < 0.05). Hematoxylin-eosin-stained sections showed bony union and increased bone trabecula in B and C groups and areas with particles positive for alkaline phosphatase staining were more abundant in groups B and C, especially in group C. CONCLUSION Neural regulation plays an important role in fracture healing. Treatment with BMSCs has a positive effect on defective callus in rats that have been subjected to SCT.
Collapse
Affiliation(s)
- Shuan-Hu Lei
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Wang Y, Yuan M, Guo QY, Lu SB, Peng J. Mesenchymal Stem Cells for Treating Articular Cartilage Defects and Osteoarthritis. Cell Transplant 2014; 24:1661-78. [PMID: 25197793 DOI: 10.3727/096368914x683485] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Articular cartilage damage and osteoarthritis are the most common joint diseases. Joints are prone to damage caused by sports injuries or aging, and such damage regularly progresses to more serious joint disorders, including osteoarthritis, which is a degenerative disease characterized by the thinning and eventual wearing out of articular cartilage, ultimately leading to joint destruction. Osteoarthritis affects millions of people worldwide. Current approaches to repair of articular cartilage damage include mosaicplasty, microfracture, and injection of autologous chondrocytes. These treatments relieve pain and improve joint function, but the long-term results are unsatisfactory. The long-term success of cartilage repair depends on development of regenerative methodologies that restore articular cartilage to a near-native state. Two promising approaches are (i) implantation of engineered constructs of mesenchymal stem cell (MSC)-seeded scaffolds, and (ii) delivery of an appropriate population of MSCs by direct intra-articular injection. MSCs may be used as trophic producers of bioactive factors initiating regenerative activities in a defective joint. Current challenges in MSC therapy are the need to overcome current limitations in cartilage cell purity and to in vitro engineer tissue structures exhibiting the required biomechanical properties. This review outlines the current status of MSCs used in cartilage tissue engineering and in cell therapy seeking to repair articular cartilage defects and related problems. MSC-based technologies show promise when used to repair cartilage defects in joints.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | |
Collapse
|
71
|
Amable PR, Teixeira MVT, Carias RBV, Granjeiro JM, Borojevic R. Protein synthesis and secretion in human mesenchymal cells derived from bone marrow, adipose tissue and Wharton's jelly. Stem Cell Res Ther 2014; 5:53. [PMID: 24739658 PMCID: PMC4055160 DOI: 10.1186/scrt442] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/01/2014] [Indexed: 02/07/2023] Open
Abstract
Introduction Different mesenchymal stromal cells (MSC) have been successfully isolated and expanded in vitro and nowadays they are tested in clinical trials for a wide variety of diseases. Whether all MSC express the same cell surface markers or have a similar secretion profile is still controversial, making it difficult to decide which stromal cell may be better for a particular application. Methods We isolated human mesenchymal stromal cells from bone marrow (BM), adipose tissue (AT) and Wharton’s jelly (WJ) and cultured them in fetal bovine serum supplemented media. We evaluated proliferation, in vitro differentiation (osteogenic, adipogenic and chondrogenic potential), expression of cell surface markers and protein secretion using Luminex and ELISA assays. Results Cell proliferation was higher for WJ-MSC, followed by AT-MSC. Differences in surface expression markers were observed only for CD54 and CD146. WJ-MSC secreted higher concentrations of chemokines, pro-inflammatory proteins and growth factors. AT-MSC showed a better pro-angiogenic profile and secreted higher amounts of extracellular matrix components and metalloproteinases. Conclusions Mesenchymal stromal cells purified from different tissues have different angiogenic, inflammatory and matrix remodeling potential properties. These abilities should be further characterized in order to choose the best protocols for their therapeutic use.
Collapse
|
72
|
Ingavle GC, Gehrke SH, Detamore MS. The bioactivity of agarose-PEGDA interpenetrating network hydrogels with covalently immobilized RGD peptides and physically entrapped aggrecan. Biomaterials 2014; 35:3558-70. [PMID: 24462353 PMCID: PMC3936106 DOI: 10.1016/j.biomaterials.2014.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/01/2014] [Indexed: 10/25/2022]
Abstract
Our previous reports of interpenetrating networks (IPNs) have demonstrated drastic improvements in mechanical performance relative to individual constituent networks while maintaining viability of encapsulated cells. The current study investigated whether covalent linkage of RGD to the poly(ethylene glycol) diacrylate (PEGDA) network could improve upon cell viability and performance of agarose-PEGDA IPNs compared to unmodified IPNs (control) and to IPNs with different concentrations of physically entrapped aggrecan, providing a point of comparison to previous work. The inclusion of RGD or aggrecan generally did not adversely affect mechanical performance, and significantly improved chondrocyte viability and performance. Although both 4 and 100 μg/mL of aggrecan improved cell viability, only 100 μg/mL aggrecan was clearly beneficial to improving biosynthesis, whereas 100 μg/mL of RGD was beneficial to both chondrocyte viability and biosynthesis. Interestingly, clustering of cells within the IPNs with RGD and the higher aggrecan concentration were observed, likely indicating cell migration and/or preferred regional proliferation. This clustering resulted in a clearly visible enhancement of matrix production compared to the other IPNs. With this cell migration, we also observed significant cell proliferation and matrix synthesis beyond the periphery of the IPN, which could have important implications in facilitating integration with surrounding cartilage in vivo. With RGD and aggrecan (at its higher concentration) providing substantial and comparable improvements in cell performance, RGD would be the recommended bioactive signal for this particular IPN formulation and cell source given the significant cost savings and potentially more straightforward regulatory pathway in commercialization.
Collapse
Affiliation(s)
- Ganesh C Ingavle
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045-7609, USA.
| | - Stevin H Gehrke
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045-7609, USA.
| | - Michael S Detamore
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045-7609, USA.
| |
Collapse
|
73
|
Co C, Vickaryous MK, Koch TG. Membrane culture and reduced oxygen tension enhances cartilage matrix formation from equine cord blood mesenchymal stromal cells in vitro. Osteoarthritis Cartilage 2014; 22:472-80. [PMID: 24418676 DOI: 10.1016/j.joca.2013.12.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/08/2013] [Accepted: 12/20/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Ongoing research is aimed at increasing cartilage tissue yield and quality from multipotent mesenchymal stromal cells (MSC) for the purpose of treating cartilage damage in horses. Low oxygen culture has been shown to enhance chondrogenesis, and novel membrane culture has been proposed to increase tissue yield and homogeneity. The objective of this study was to evaluate and compare the effect of reduced oxygen and membrane culture during in vitro chondrogenesis of equine cord blood (CB) MSC. METHODS CB-MSC (n = 5 foals) were expanded at 21% oxygen prior to 3-week differentiation in membrane or pellet culture at 5% and 21% oxygen. Assessment included histological examination (H&E, toluidine Blue, immunohistochemistry (IHC) for collagen type I and II), protein quantification by hydroxyproline assay and dimethylmethylene assay, and mRNA analysis for collagen IA1, collagen IIA1, collagen XA1, HIF1α and Sox9. RESULTS Among treatment groups, 5% membrane culture produced neocartilage most closely resembling hyaline cartilage. Membrane culture resulted in increased wet mass, homogenous matrix morphology and an increase in total collagen content, while 5% oxygen culture resulted in higher GAG and type II collagen content. No significant differences were observed for mRNA analysis. CONCLUSION Membrane culture at 5% oxygen produces a comparatively larger amount of higher quality neocartilage. Matrix homogeneity is attributed to a uniform diffusion gradient and reduced surface tension. Membrane culture holds promise for scale-up for therapeutic purposes, for cellular preconditioning prior to cytotherapeutic applications, and for modeling system for gas-dependent chondrogenic differentiation studies.
Collapse
Affiliation(s)
- C Co
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - M K Vickaryous
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - T G Koch
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; Department of Clinical Medicine, Orthopaedic Research Laboratory, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
74
|
Mellott AJ, Godsey ME, Shinogle HE, Moore DS, Forrest ML, Detamore MS. Improving viability and transfection efficiency with human umbilical cord wharton's jelly cells through use of a ROCK inhibitor. Cell Reprogram 2014; 16:91-7. [PMID: 24552552 DOI: 10.1089/cell.2013.0069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Differentiating stem cells using gene delivery is a key strategy in tissue engineering and regenerative medicine applications. Nonviral gene delivery bypasses several safety concerns associated with viral gene delivery; however, leading nonviral techniques, such as electroporation, subject cells to high stress and can result in poor cell viabilities. Inhibition of Rho-associated coiled-coil kinase (ROCK) has been shown to mitigate apoptotic mechanisms associated with detachment and freezing of induced pluripotent stem cells and embryonic stem cells; however, inhibiting ROCK in mesenchymal stromal cells (MSCs) for improving gene delivery applications has not been reported previously. In this study, we hypothesized that ROCK Inhibitor (RI) would improve cell viability and gene expression in primary human umbilical cord mesenchymal stromal cells (hUCMSCs) when transfected via Nucleofection™. As hypothesized, the pre-treatment and post-treatment of hUCMSCs transfected via nucleofection with Y-27632-RI significantly improved survival rates of hUCMSCs and gene expression as measured by green fluorescent protein intensity. This study provides the first comparative look at the effect of Y-27632-RI on hUCMSCs that underwent transfection via nucleofection and shows that using Y-27632-RI in concert with nucleofection could greatly enhance the utility of differentiating and reprogramming hUCMSCs for tissue engineering applications.
Collapse
Affiliation(s)
- Adam J Mellott
- 1 Bioengineering Program, University of Kansas , Lawrence, KS, 66045
| | | | | | | | | | | |
Collapse
|
75
|
Granéli C, Thorfve A, Ruetschi U, Brisby H, Thomsen P, Lindahl A, Karlsson C. Novel markers of osteogenic and adipogenic differentiation of human bone marrow stromal cells identified using a quantitative proteomics approach. Stem Cell Res 2014; 12:153-65. [DOI: 10.1016/j.scr.2013.09.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 09/17/2013] [Accepted: 09/19/2013] [Indexed: 12/22/2022] Open
|
76
|
Detamore MS. Human umbilical cord mesenchymal stromal cells in regenerative medicine. Stem Cell Res Ther 2013; 4:142. [PMID: 24456755 PMCID: PMC4029508 DOI: 10.1186/scrt353] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cells of the human umbilical cord offer tremendous potential for improving human health. Cells from the Wharton’s jelly (umbilical cord stroma) in particular, referred to as human umbilical cord mesenchymal stromal cells (HUCMSCs), hold several advantages that make them appealing for translational research. In the previous issue of Stem Cell Research & Therapy, Chon and colleagues made an important contribution to the HUCMSC literature not only by presenting HUCMSCs as an emerging cell source for intervertebral disc regeneration in general and the nucleus pulposus in particular, but also by demonstrating that an extracellular matrix-based strategy might be preferred over the use of growth factors. By culturing HUCMSCs under hypoxia in serum-free conditions in the presence of Matrigel with laminin-111, they were able to achieve intense collagen II staining by 21 days without the addition of exogenous growth factors. There is tremendous translational significance here in that such raw materials may alleviate the need for the use of growth factors in some instances, and this may have important ramifications in reducing product cost and streamlining regulatory approval. Chon and colleagues provide a promising example of the potential of HUCMSCs, demonstrating the ability to guide HUCMSC differentiation even in the absence of serum and growth factors and supporting the use of HUCMSCs as a viable alternative in intervertebral disc regeneration.
Collapse
|
77
|
Alves da Silva ML, Costa-Pinto AR, Martins A, Correlo VM, Sol P, Bhattacharya M, Faria S, Reis RL, Neves NM. Conditioned medium as a strategy for human stem cells chondrogenic differentiation. J Tissue Eng Regen Med 2013; 9:714-23. [PMID: 24155167 DOI: 10.1002/term.1812] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 07/24/2013] [Indexed: 02/04/2023]
Abstract
Paracrine signalling from chondrocytes has been reported to increase the synthesis and expression of cartilage extracellular matrix (ECM) by stem cells. The use of conditioned medium obtained from chondrocytes for stimulating stem cells chondrogenic differentiation may be a very interesting alternative for moving into the clinical application of these cells, as chondrocytes could be partially replaced by stem cells for this type of application. In the present study we aimed to achieve chondrogenic differentiation of two different sources of stem cells using conditioned medium, without adding growth factors. We tested both human bone marrow-derived mesenchymal stem cells (hBSMCs) and human Wharton's jelly-derived stem cells (hWJSCs). Conditioned medium obtained from a culture of human articular chondrocytes was used to feed the cells during the experiment. Cultures were performed in previously produced three-dimensional (3D) scaffolds, composed of a blend of 50:50 chitosan:poly(butylene succinate). Both types of stem cells were able to undergo chondrogenic differentiation without the addition of growth factors. Cultures using hWJSCs showed significantly higher GAGs accumulation and expression of cartilage-related genes (aggrecan, Sox9 and collagen type II) when compared to hBMSCs cultures. Conditioned medium obtained from articular chondrocytes induced the chondrogenic differentiation of MSCs and ECM formation. Obtained results showed that this new strategy is very interesting and should be further explored for clinical applications.
Collapse
Affiliation(s)
- M L Alves da Silva
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - A R Costa-Pinto
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - A Martins
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - V M Correlo
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - P Sol
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - M Bhattacharya
- Department of Biosystems Engineering, University of Minnesota, Minneapolis, MN, USA
| | - S Faria
- CMAT, Mathematical Research Centre, Department of Mathematics and Applications, University of Minho, Guimarães, Portugal
| | - R L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| | - Nuno M Neves
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's, PT Government Associated Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
78
|
Chick stem cells: current progress and future prospects. Stem Cell Res 2013; 11:1378-92. [PMID: 24103496 PMCID: PMC3989061 DOI: 10.1016/j.scr.2013.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 09/06/2013] [Accepted: 09/13/2013] [Indexed: 12/15/2022] Open
Abstract
Chick embryonic stem cells (cESCs) can be derived from cells obtained from stage X embryos (blastoderm stage); these have the ability to contribute to all somatic lineages in chimaeras, but not to the germ line. However, lines of stem cells that are able to contribute to the germ line can be established from chick primordial germ cells (cPGCs) and embryonic germ cells (cEGCs). This review provides information on avian stem cells, emphasizing different sources of cells and current methods for derivation and culture of pluripotent cells from chick embryos. We also review technologies for isolation and derivation of chicken germ cells and the production of transgenic birds. Chick embryonic stem cells (cESCs) can be derived from a variety of sources. cESCs can contribute to all somatic cell types but not to the germ line. germ cells can be isolated from early embryos, embryonic blood and gonads. germ cells can establish self-renewing lines and contribute to the germline.
Collapse
|
79
|
Wang L, Weiss ML, Detamore MS. Recent Patents Pertaining to Immune Modulation and Musculoskeletal Regeneration with Wharton's Jelly Cells. ACTA ACUST UNITED AC 2013; 3:182-192. [PMID: 26279972 DOI: 10.2174/22102965113039990020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Umbilical cord mesenchymal stromal cells (UCMSCs) are isolated from Wharton's jelly in the umbilical cord at birth, and offer advantages over adult mesenchymal stromal cells (MSCs) such as highly efficient isolation, faster proliferation in vitro, a broader differentiation potential, and non-invasive harvesting procedure. Their expansion and differentiation potential renders them a promising cell source for tissue engineering and clinical applications. This review discusses recent updates on the differentiation strategies for musculoskeletal tissue engineering including cartilage, bone, and muscle. In addition to tissue engineering applications, UCMSCs can be utilized to support hematopoiesis and modulate immune response. We review the patents relevant to the application of MSCs including UCMSCs in hematopoiesis and immune modulation. Finally, the current hurdles in the clinical translation of UCMSCs are discussed. During clinical translation, it is critical to develop large-scale manufacturing of UCMSCs as well as the composition of expansion and differentiation media. Four clinical trials to date have examined the safety and efficacy of UCMSCs. Once public banking of UCMSCs is available to supply matched allogeneic units and once UCMSC manufacturing is standardized, we anticipate that UCMSCs will be more widely used in clinical trials.
Collapse
Affiliation(s)
- Limin Wang
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas 66506, USA
| | - Michael S Detamore
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, USA
| |
Collapse
|
80
|
Liu S, Hou KD, Yuan M, Peng J, Zhang L, Sui X, Zhao B, Xu W, Wang A, Lu S, Guo Q. Characteristics of mesenchymal stem cells derived from Wharton's jelly of human umbilical cord and for fabrication of non-scaffold tissue-engineered cartilage. J Biosci Bioeng 2013; 117:229-235. [PMID: 23899897 DOI: 10.1016/j.jbiosc.2013.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 06/06/2013] [Accepted: 07/01/2013] [Indexed: 12/22/2022]
Abstract
Once cartilage is damaged, it has limited potential for self-repair. Autologous chondrocyte implantation is an effective treatment, but patients may suffer during cartilage harvesting and the donor-site morbidity may accelerate joint degeneration. Using autologous mesenchymal stem cells (MSCs) derived chondrocytes is another selection, while it also causes some injuring. The umbilical cord, an ecto-embryo tissue may be an ideal source of cells, because of its accessibility, abundant resources, painless procedures for harvesting, and lack of ethical issues. We isolated MSCs from Wharton's jelly of human umbilical cord (WMSCs), which expressed CD44, CD105 and CD271 but not CD34 and CD45 with flow cytometry analysis. RT-PCR showed not only positive expression of CD90, c-kit, Sca1, SH2 and SH3 but also positive expression of the chondrocyte markers Sox-9 and Col-2A1. WMSCs cultured in high-density in the presence of transforming growth factor β1 and dexamethasone showed cartilage extracellular matrix-secretion and integrated into a thin piece of cell-based membrane. The cell-based thin membrane cultured in rotary cell culture system formed a round, opaque, glistening non-scaffold cartilage-like tissue, larger and condenser than what was formed with conventional pellet culture. Glycosaminoglycan and type II collagen content significantly increased after 3-week culture. The human WMSCs express characteristics of pre-chondrocytes, low immunogenicity and are easy to be obtained with higher purity because there have no hematopoietic cells in Wharton's jelly, so it may be a new seed cells more suitable for constructing tissue-engineered cartilage.
Collapse
Affiliation(s)
- Shuyun Liu
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Ke Dong Hou
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Mei Yuan
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Jiang Peng
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Li Zhang
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Xiang Sui
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Bin Zhao
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Wenjing Xu
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Aiyuan Wang
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Shibi Lu
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | - Quanyi Guo
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China.
| |
Collapse
|
81
|
Corotchi MC, Popa MA, Remes A, Sima LE, Gussi I, Lupu Plesu M. Isolation method and xeno-free culture conditions influence multipotent differentiation capacity of human Wharton's jelly-derived mesenchymal stem cells. Stem Cell Res Ther 2013; 4:81. [PMID: 23845279 PMCID: PMC3854854 DOI: 10.1186/scrt232] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 07/08/2013] [Indexed: 01/08/2023] Open
Abstract
Introduction Human Wharton’s jelly (WJ) has become a preferred source of mesenchymal stem cells (MSCs) whose clinical applications are limited by the use of adequate xeno-free (XF), in vitro manipulation conditions. Therefore, the objective of our study was to characterize WJ-derived MSCs (WJ-MSCs), isolated by different methods and cultured in a commercially available, MSC XF medium, not least of all by investigating their endothelial differentiation capacity. Methods WJ explants and enzymatically dissociated WJ cells were cultured in a defined, XF medium for MSCs. Adherent cells at passages 2 and 5 were characterized as MSCs by flow cytometry, MTT, real-time quantitative reverse transcription PCR, and functional multipotent differentiation assays. The endothelial differentiation capacity of MSCs isolated and expanded until passage 2 in the MSC XF medium, and then subcultured for five passages in a commercially available endothelial growth medium (group A), was assessed over serial passages, as compared to adherent WJ-derived cells isolated and expanded for five consecutive passages in the endothelial medium (group B). Results The MSC phenotype of WJ explant- and pellet-derived cells, isolated and expanded in the MSC XF medium, was proven based on the expression of CD44/CD73/CD90/CD105 surface markers and osteo-/adipo-/chondrogenic multipotent differentiation potential, which differed according to the isolation method and/or passage number. Upon exposure to endothelial differentiation cues, cells belonging to group A did not exhibit endothelial cell characteristics over serial passages; by contrast, WJ pellet-derived cells belonging to group B expressed endothelial characteristics at gene, protein and functional levels, potentially due to culture conditions favoring the isolation of other stem/progenitor cell types than MSCs, able to give rise to an endothelial progeny. Conclusions The use of defined, MSC XF media for isolation and expansion of human WJ-MSCs is a prerequisite for the establishment of their real endothelial differentiation capacity, as candidates for clinical therapy applications. Thus, the standardization of WJ-MSCs isolation and culture expansion techniques in defined, MSC XF media, for their accurate characterization, would be a priority in the stem cell research field.
Collapse
|
82
|
Side-by-side comparison of the biological characteristics of human umbilical cord and adipose tissue-derived mesenchymal stem cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:438243. [PMID: 23936800 PMCID: PMC3722850 DOI: 10.1155/2013/438243] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/20/2022]
Abstract
Both human adipose tissue-derived mesenchymal stem cells (ASCs) and umbilical cord-derived mesenchymal stem cells (UC-MSCs) have been explored as attractive mesenchymal stem cells (MSCs) sources, but very few parallel comparative studies of these two cell types have been made. We designed a side-by-side comparative study by isolating MSCs from the adipose tissue and umbilical cords from mothers delivering full-term babies and thus compared the various biological aspects of ASCs and UC-MSCs derived from the same individual, in one study. Both types of cells expressed cell surface markers characteristic of MSCs. ASCs and UC-MSCs both could be efficiently induced into adipocytes, osteoblasts, and neuronal phenotypes. While there were no significant differences in their osteogenic differentiation, the adipogenesis of ASCs was more prominent and efficient than UC-MSCs. In the meanwhile, ASCs responded better to neuronal induction methods, exhibiting the higher differentiation rate in a relatively shorter time. In addition, UC-MSCs exhibited a more prominent secretion profile of cytokines than ASCs. These results indicate that although ASCs and UC-MSCs share considerable similarities in their immunological phenotype and pluripotentiality, certain biological differences do exist, which might have different implications for future cell-based therapy.
Collapse
|
83
|
Yang W, Li Z, Zhang W, Luo C, Ouyang Q, Yang G, Wang Y. A novel density control device for the study of cancer cell autocrine effect. Biomed Microdevices 2013; 15:683-689. [PMID: 23784259 DOI: 10.1007/s10544-013-9783-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this paper, we propose a novel cell self-loading and patterning device for quantitatively study density effect on cell behaviors. Using this device, it is easy to gather different cell density colonies in different sizes of micro-chambers using one homogeneous cell solution. As a demonstration, we show that the cell number of self-patterning MCF-7 colony is in proportion to the size of liquid-absorbing cavity in the device, from single cell to tens of cells. This device can easily be used to compare the cancer cells' proliferation in different micro-environments, such as the same number of cells in micro-cavities with different sizes, or different numbers of cells in micro-cavities with the same size, or with different FBS concentrations. Our studies imply a plausible positive correlation between the local concentration of autocrine factors and tumor cell proliferation, which is also quantitative analyzed by a simple model.
Collapse
Affiliation(s)
- Wei Yang
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, People's Republic of China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China
| | - Zhaojun Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China
| | - Weilin Zhang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, People's Republic of China.
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China.
| | - Qi Ouyang
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, People's Republic of China.
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, People's Republic of China.
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, Peking University, Beijing, 100871, People's Republic of China
| | - Yugang Wang
- State Key Laboratory of Nuclear Physics and Technology, Peking University, Beijing, 100871, People's Republic of China
| |
Collapse
|
84
|
Differentiation of mesenchymal stem cells from human umbilical cord tissue into odontoblast-like cells using the conditioned medium of tooth germ cells in vitro. BIOMED RESEARCH INTERNATIONAL 2013; 2013:218543. [PMID: 23762828 PMCID: PMC3666309 DOI: 10.1155/2013/218543] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/08/2013] [Indexed: 12/31/2022]
Abstract
The easily accessible mesenchymal stem cells in the Wharton's jelly of human umbilical cord tissue (hUCMSCs) have excellent proliferation and differentiation potential, but it remains unclear whether hUCMSCs can differentiate into odontoblasts. In this study, mesenchymal stem cells were isolated from the Wharton's jelly of human umbilical cord tissue using the simple method of tissue blocks culture attachment. UCMSC surface marker expression was then evaluated for the isolated cells using flow cytometry. The third-passage hUCMSCs induced by conditioned medium from developing tooth germ cells (TGC-CM) displayed high alkaline phosphatase (ALP) levels (P < 0.001), an enhanced ability to proliferate (P < 0.05), and the presence of mineralized nodules. These effects were not observed in cells treated with regular medium. After induction of hUCMSCs, the results of reverse transcriptional polymerase chain reaction (PCR) indicated that the dentin sialophosphoprotein (DSPP) and dentin matrix protein 1 (DMP1) genes were significantly tested. Additionally, dentin sialoprotein (DSP) and DMP1 demonstrated significant levels of staining in an immunofluorescence analysis. In contrast, the control cells failed to display the characteristics of odontoblasts. Taken together, these results suggest that hUCMSCs can be induced to differentiate into odontoblast-like cells with TGC-CM and provide a novel strategy for tooth regeneration research.
Collapse
|
85
|
Ingavle GC, Frei AW, Gehrke SH, Detamore MS. Incorporation of aggrecan in interpenetrating network hydrogels to improve cellular performance for cartilage tissue engineering. Tissue Eng Part A 2013; 19:1349-59. [PMID: 23379843 DOI: 10.1089/ten.tea.2012.0160] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Interpenetrating network (IPN) hydrogels were recently introduced to the cartilage tissue engineering literature, with the approach of encapsulating cells in thermally gelling agarose that is then soaked in a poly(ethylene glycol) diacrylate (PEGDA) solution, which is then photopolymerized. These IPNs possess significantly enhanced mechanical performance desirable for cartilage regeneration, potentially allowing patients to return to weight-bearing activities quickly after surgical implantation. In an effort to improve cell viability and performance, inspiration was drawn from previous studies that have elicited positive chondrogenic responses to aggrecan, the proteoglycan largely responsible for the compressive stiffness of cartilage. Aggrecan was incorporated into the IPNs in conservative concentrations (40 μg/mL), and its effect was contrasted with the incorporation of chondroitin sulfate (CS), the primary glycosaminoglycan associated with aggrecan. Aggrecan was incorporated by physical entrapment within agarose and methacrylated CS was incorporated by copolymerization with PEGDA. The IPNs incorporating aggrecan or CS exhibited over 50% viability with encapsulated chondrocytes after 6 weeks. Both aggrecan and CS improved cell viability by 15.6% and 20%, respectively, relative to pure IPNs at 6 weeks culture time. In summary, we have introduced the novel approach of including a raw material from cartilage, namely aggrecan, to serve as a bioactive signal to cells encapsulated in IPN hydrogels for cartilage tissue engineering, which led to improved performance of encapsulated chondrocytes.
Collapse
Affiliation(s)
- Ganesh C Ingavle
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045-7609, USA
| | | | | | | |
Collapse
|
86
|
Li SL, Liu Y, Hui L. Construction of engineering adipose-like tissue in vivo utilizing human insulin gene-modified umbilical cord mesenchymal stromal cells with silk fibroin 3D scaffolds. J Tissue Eng Regen Med 2013; 9:E267-75. [PMID: 23509085 DOI: 10.1002/term.1695] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 12/19/2012] [Indexed: 11/12/2022]
Abstract
We evaluated the use of a combination of human insulin gene-modified umbilical cord mesenchymal stromal cells (hUMSCs) with silk fibroin 3D scaffolds for adipose tissue engineering. In this study hUMSCs were isolated and cultured. HUMSCs infected with Ade-insulin-EGFP were seeded in fibroin 3D scaffolds with uniform 50-60 µm pore size. Silk fibroin scaffolds with untransfected hUMSCs were used as control. They were cultured for 4 days in adipogenic medium and transplanted under the dorsal skins of female Wistar rats after the hUMSCs had been labelled with chloromethylbenzamido-1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (CM-Dil). Macroscopical impression, fluorescence observation, histology and SEM were used for assessment after transplantation at 8 and 12 weeks. Macroscopically, newly formed adipose tissue was observed in the experimental group and control group after 8 and 12 weeks. Fluorescence observation supported that the formed adipose tissue originated from seeded hUMSCs rather than from possible infiltrating perivascular tissue. Oil red O staining of newly formed tissue showed that there was substantially more tissue regeneration in the experimental group than in the control group. SEM showed that experimental group cells had more fat-like cells, whose volume was larger than that of the control group, and degradation of the silk fibroin scaffold was greater under SEM observation. This study provides significant evidence that hUMSCs transfected by adenovirus vector have good compatibility with silk fibroin scaffold, and adenoviral transfection of the human insulin gene can be used for the construction of tissue-engineered adipose.
Collapse
Affiliation(s)
- Shi-Long Li
- Burns and Plastic Surgery Centre, Lanzhou General Hospital of Lanzhou Command of CPLA, People's Republic of China
| | - Yi Liu
- Burns and Plastic Surgery Centre, Lanzhou General Hospital of Lanzhou Command of CPLA, People's Republic of China
| | - Ling Hui
- Key Laboratory of Stem Cells and Gene Drugs, Lanzhou General Hospital of Lanzhou Command of CPLA, People's Republic of China
| |
Collapse
|
87
|
Gibbons MC, Foley MA, Cardinal KO. Thinking inside the box: keeping tissue-engineered constructs in vitro for use as preclinical models. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:14-30. [PMID: 22800715 DOI: 10.1089/ten.teb.2012.0305] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue engineers have made great strides toward the creation of living tissue replacements for a wide range of tissue types and applications, with eventual patient implantation as the primary goal. However, an alternate use of tissue-engineered constructs exists: as in vitro preclinical models for purposes such as drug screening and device testing. Tissue-engineered preclinical models have numerous potential advantages over existing models, including cultivation in three-dimensional geometries, decreased cost, increased reproducibility, precise control over cultivation conditions, and the incorporation of human cells. Over the past decade, a number of researchers have developed and used tissue-engineered constructs as preclinical models for testing pharmaceuticals, gene therapies, stents, and other technologies, with examples including blood vessels, skeletal muscle, bone, cartilage, skin, cardiac muscle, liver, cornea, reproductive tissues, adipose, small intestine, neural tissue, and kidney. The focus of this article is to review accomplishments toward the creation and use of tissue-engineered preclinical models of each of these different tissue types.
Collapse
Affiliation(s)
- Michael C Gibbons
- Department of Biomedical and General Engineering, Cal Poly San Luis Obispo, San Luis Obispo, California 93407, USA
| | | | | |
Collapse
|
88
|
Therapeutic application of mesenchymal stem cells in bone and joint diseases. Clin Exp Med 2012; 14:13-24. [DOI: 10.1007/s10238-012-0218-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 10/22/2012] [Indexed: 02/06/2023]
|
89
|
A new era of cartilage repair using cell therapy and tissue engineering: turning current clinical limitations into new ideas. Tissue Eng Regen Med 2012. [DOI: 10.1007/s13770-012-0370-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
90
|
Beane OS, Darling EM. Isolation, characterization, and differentiation of stem cells for cartilage regeneration. Ann Biomed Eng 2012; 40:2079-97. [PMID: 22907257 DOI: 10.1007/s10439-012-0639-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/08/2012] [Indexed: 12/27/2022]
Abstract
The goal of tissue engineering is to create a functional replacement for tissues damaged by injury or disease. In many cases, impaired tissues cannot provide viable cells, leading to the investigation of stem cells as a possible alternative. Cartilage, in particular, may benefit from the use of stem cells since the tissue has low cellularity and cannot effectively repair itself. To address this need, researchers are investigating the chondrogenic capabilities of several multipotent stem cell sources, including adult and extra-embryonic mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). Comparative studies indicate that each cell type has advantages and disadvantages, and while direct comparisons are difficult to make, published data suggest some sources may be more promising for cartilage regeneration than others. In this review, we identify current approaches for isolating and chondrogenically differentiating MSCs from bone marrow, fat, synovium, muscle, and peripheral blood, as well as cells from extra-embryonic tissues, ESCs, and iPSCs. Additionally, we assess chondrogenic induction with growth factors, identifying standard cocktails used for each stem cell type. Cell-only (pellet) and scaffold-based studies are also included, as is a discussion of in vivo results.
Collapse
Affiliation(s)
- Olivia S Beane
- Center for Biomedical Engineering, Brown University, Providence, RI, USA
| | | |
Collapse
|
91
|
Wen Y, Jiang B, Cui J, Li G, Yu M, Wang F, Zhang G, Nan X, Yue W, Xu X, Pei X. Superior osteogenic capacity of different mesenchymal stem cells for bone tissue engineering. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 116:e324-32. [PMID: 22841430 DOI: 10.1016/j.oooo.2012.02.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 01/18/2012] [Accepted: 02/07/2012] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We evaluated the effect of human bone marrow stromal cells (hBMSCs), human adipose tissue-derived mesenchymal stem cells (hAD-MSCs), and umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in bone tissue engineering and identified a reliable cell source. STUDY DESIGN Alkaline phosphatase (ALP) activity and quantitative polymerase chain reaction were used to evaluate osteogenic in vitro, X-ray and histologic analysis in vivo. RESULTS hBMSCs exhibited strongest ALP staining, followed by hAD-MSCs and hUC-MSCs. At 7 days, hUC-MSCs and hAD-MSCs had higher expression of collagen type I and Runt-related transcription factor 2 than hBMSCs, and hUC-MSCs showed higher osteopontin expression. Bone structure was observed in the hUC-MSC group. Defects showed good healing in the hBMSC and hAD-MSC groups. Enhanced green fluorescent protein and osteopontin were detected in newly formed bone at 8 weeks. CONCLUSIONS Our results suggested that hUC-MSCs and hAD-MSCs could be used for bone tissue engineering effectively; hUC-MSCs could serve as a new alternative cell source.
Collapse
Affiliation(s)
- Yong Wen
- School of Stomatology, Shandong University, Jinan, China; Stem Cell and Regenerative Medicine Laboratory, Beijing Institute of Transfusion Medicine, Beijing, China; Shandong Provincial Key Laboratory of Oral Biomedicine, Jinan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Liu J, Zhou H, Weir MD, Xu HHK, Chen Q, Trotman CA. Fast-degradable microbeads encapsulating human umbilical cord stem cells in alginate for muscle tissue engineering. Tissue Eng Part A 2012; 18:2303-14. [PMID: 22697426 DOI: 10.1089/ten.tea.2011.0658] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells (hUCMSCs) are inexhaustible and can be obtained without an invasive surgery. To date, there has been no report on seeding hUCMSCs in three-dimensional scaffolds for muscle tissue engineering. The objectives of this study were to (1) investigate hUCMSC seeding in a scaffold for muscle engineering and (2) develop a novel construct consisting of hUCMSC-encapsulating and fast-degradable microbeads inside a hydrogel matrix. The rationale was that the hydrogel matrix would maintain the defect volume, while the microbeads would degrade to release the cells and concomitantly create macropores in the matrix. hUCMSCs were encapsulated in alginate-fibrin microbeads, which were packed in an Arg-Gly-Asp (RGD)-modified alginate matrix (AM). This construct is referred to as hUCMSC-microbead-AM. The control consisted of the usual cell encapsulation in AM without microbeads (referred to as hUCMSC-AM). In the hUCMSC-AM construct, the hUCMSCs showed as round dots with no spreading at 1-14 days. In contrast, cells in the hUCMSC-microbead-AM construct had a healthy spreading and elongated morphology. The microbeads successfully degraded and released the cells at 8 days. Myogenic expressions for hUCMSC-microbead-AM were more than threefold those of hUCMSC-AM (p<0.05). Immunofluorescence for myogenic markers was much stronger for hUCMSC-microbead-AM than hUCMSC-AM. Muscle creatine kinase of hUCMSC-microbead-AM at 14 days was twofold that of hUCMSC-AM (p<0.05). In conclusion, hUCMSC encapsulation in novel fast-degradable microbeads inside a hydrogel matrix was investigated for muscle engineering. Compared to the usual method of seeding cells in a hydrogel matrix, hUCMSC-microbead-AM construct had greatly improved cell viability and myogenic differentiation, and hence, is promising to enhance muscle regeneration.
Collapse
Affiliation(s)
- Jun Liu
- Biomaterials and Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, Maryland 21201, USA
| | | | | | | | | | | |
Collapse
|
93
|
Fong CY, Subramanian A, Gauthaman K, Venugopal J, Biswas A, Ramakrishna S, Bongso A. Human umbilical cord Wharton's jelly stem cells undergo enhanced chondrogenic differentiation when grown on nanofibrous scaffolds and in a sequential two-stage culture medium environment. Stem Cell Rev Rep 2012; 8:195-209. [PMID: 21671058 DOI: 10.1007/s12015-011-9289-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current treatments used for osteoarthritis from cartilage damage have their disadvantages of donor site morbidity, complicated surgical interventions and risks of infection and graft rejection. Recent advances in tissue engineering have offered much promise in cartilage repair but the best cell source and in vitro system have not as yet been optimised. Human bone marrow mesenchymal stem cells (hBMSCs) have thus far been the cell of choice. However, we derived a unique stem cell from the human umbilical cord Wharton's jelly (hWJSC) that has properties superior to hBMSCs in terms of ready availability, prolonged stemness characteristics in vitro, high proliferation rates, wide multipotency, non-tumorigenicity and tolerance in allogeneic transplantation. We observed enhanced cell attachment, cell proliferation and chondrogenesis of hWJSCs over hBMSCs when grown on PCL/Collagen nanoscaffolds in the presence of a two-stage sequential complex/chondrogenic medium for 21 days. Improvement of these three parameters were confirmed via inverted optics, field emission scanning electron microscopy (FESEM), MTT assay, pellet diameters, Alcian blue histology and staining, glycosaminglycans (GAG) and hyaluronic acid production and expression of key chondrogenic genes (SOX9, Collagen type II, COMP, FMOD) using immunohistochemistry and real-time polymerase chain reaction (qRT-PCR). In separate experiments we demonstrated that the 16 ng/ml of basic fibroblast growth factor (bFGF) present in the complex medium may have contributed to driving chondrogenesis. We conclude that hWJSCs are an attractive stem cell source for inducing chondrogenesis in vitro when grown on nanoscaffolds and exposed sequentially first to complex medium and then followed by chondrogenic medium.
Collapse
Affiliation(s)
- Chui-Yee Fong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | | | | | | | | | | | | |
Collapse
|
94
|
Lui PPY, Chan KM. Tendon-derived stem cells (TDSCs): from basic science to potential roles in tendon pathology and tissue engineering applications. Stem Cell Rev Rep 2012; 7:883-97. [PMID: 21611803 DOI: 10.1007/s12015-011-9276-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traditionally, tendons are considered to only contain tenocytes that are responsible for the maintenance, repair and remodeling of tendons. Stem cells, which are termed tendon-derived stem cells (TDSCs), have recently been identified in tendons. This review aims to summarize the current information about the in vitro characteristics of TDSCs, including issues related to TDSC isolation and culture, their cell morphology, immunophenotypes, proliferation and differentiation characteristics and senescence during in vitro passaging. The challenges in studying the functions of these cells are also discussed. The niche where TDSCs resided essentially provides signals that are conducive to the maintenance of definitive stem cell properties of TDSCs. Yet the niche may also induce pathologies by imposing an aberrant function on TDSCs or other targets. The possible niche factors of TDSCs are herein discussed. We presented current evidences supporting the potential pathogenic role of TDSCs in the development of tendinopathy with reference to the recent findings on the altered biological responses of these cells in response to their potential niche factors. The use of resident stem cells may promote engraftment and differentiation of transplanted cells in tendon and tendon-bone junction repair because the tendon milieu is an ideal and familiar environment to the transplanted cells. Evidences are presented to show the potential advantages and results of using TDSCs as a new cell source for tendon and tendon-bone junction repair. Issues pertaining to the use of TDSCs for tissue repair are also discussed.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| | | |
Collapse
|
95
|
Ingavle GC, Dormer NH, Gehrke SH, Detamore MS. Using chondroitin sulfate to improve the viability and biosynthesis of chondrocytes encapsulated in interpenetrating network (IPN) hydrogels of agarose and poly(ethylene glycol) diacrylate. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:157-70. [PMID: 22116661 PMCID: PMC3729881 DOI: 10.1007/s10856-011-4499-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 11/09/2011] [Indexed: 05/21/2023]
Abstract
We recently introduced agarose-poly(ethylene glycol) diacrylate (PEGDA) interpenetrating network (IPN) hydrogels to cartilage tissue engineering that were able to encapsulate viable cells and provide a significant improvement in mechanical performance relative to its two constituent hydrogels. The goal of the current study was to develop a novel synthesis protocol to incorporate methacrylated chondroitin sulfate (MCS) into the IPN design hypothesized to improve cell viability and biosynthesis. The IPN was formed by encapsulating porcine chondrocytes in agarose, soaking the construct in a solution of 1:10 MCS:PEGDA, which was then photopolymerized to form a copolymer network as the second network. The IPN with incorporated CS (CS-IPN) (~0.5 wt%) resulted in a 4- to 5-fold increase in the compressive elastic modulus relative to either the PEGDA or agarose gels. After 6 weeks of in vitro culture, more than 50% of the encapsulated chondrocytes remained viable within the CS-modified IPN, in contrast to 35% viability observed in the unmodified. At week 6, the CS-IPN had significantly higher normalized GAG contents (347 ± 34 μg/μg) than unmodified IPNs (158 ± 27 μg/μg, P < 0.05). Overall, the approach of incorporating biopolymers such as CS from native tissue may provide favorable micro-environment and beneficial signals to cells to enhance their overall performance in IPNs.
Collapse
|
96
|
Lavrentieva A, Hatlapatka T, Neumann A, Weyand B, Kasper C. Potential for osteogenic and chondrogenic differentiation of MSC. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 129:73-88. [PMID: 22457052 DOI: 10.1007/10_2012_133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The introduction of mesenchymal stem cells (MSC) into the field of tissue engineering for bone and cartilage repair is a promising development, since these cells can be expanded ex vivo to clinically relevant numbers and, after expansion, retain their ability to differentiate into different cell lineages. Mesenchymal stem cells isolated from various tissues have been intensively studied and characterized by many research groups. To obtain functionally active differentiated tissue, tissue engineered constructs are cultivated in vitro statically or dynamically in bioreactors under controlled conditions. These conditions include special cell culture media, addition of signalling molecules, various physical and chemical factors and the application of different mechanical stimuli. Oxygen concentration in the culture environment is also a significant factor which influences MSC proliferation, stemness and differentiation capacity. Knowledge of the different aspects which affect MSC differentiation in vivo and in vitro will help researchers to achieve directed cell fate without the addition of differentiation agents in concentrations above the physiological range.
Collapse
Affiliation(s)
- Antonina Lavrentieva
- Institut für Technische Chemie, Leibniz Universität Hannover, Callinstrasse 5, 30167, Hannover, Germany,
| | | | | | | | | |
Collapse
|
97
|
Tan Q, Lui PPY, Rui YF, Wong YM. Comparison of potentials of stem cells isolated from tendon and bone marrow for musculoskeletal tissue engineering. Tissue Eng Part A 2011; 18:840-51. [PMID: 22011320 DOI: 10.1089/ten.tea.2011.0362] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The use of tendon-derived stem cells (TDSCs) as a cell source for musculoskeletal tissue engineering has not been compared with that of bone marrow stromal cells (BMSC). This study compared the mesenchymal stem cell (MSC) and embryonic stem cells (ESC) markers, clonogenicity, proliferative capacity, and multilineage differentiation potential of rat TDSC and BMSC in vitro. The MSC and ESC marker profiles of paired TDSC and BMSC were compared using flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. Their clonogenicity and proliferative capacity were compared using colony-forming and 5-bromo-2'-deoxyuridine assays, respectively. The expression of tenogenic, osteogenic, and chondrogenic markers at basal state were examined using qRT-PCR. Their osteogenic, chondrogenic, and adipogenic differentiation potentials were compared using standard assays. TDSC and BMSC showed similar expression of CD90 and CD73. TDSC expressed higher levels of Oct4 than BMSC. TDSC exhibited higher clonogenicity, proliferated faster, and expressed higher tenomodulin, scleraxis, collagen 1 α 1 (Col1A1), decorin, alkaline phosphatase, Col2A1, and biglycan messenger RNA levels than BMSC. There was higher calcium nodule formation and osteogenic marker expression in TDSC than BMSC upon osteogenic induction. More chondrocyte-like cells and higher glycosaminoglycan deposition and chondrogenic marker expression were observed in TDSC than BMSC upon chondrogenic induction. There were more oil droplets and expression of an adipogenic marker in TDSC than BMSC upon adipogenic induction. TDSC expressed higher Oct4 levels, which was reported to positively regulate mesendodermal lineage differentiation, showed higher clonogenicity and proliferative capacity, and had greater tenogenic, osteogenic, chondrogenic, and adipogenic markers and differentiation potential than BMSC. TDSC might be a better cell source than BMSC for musculoskeletal tissue regeneration.
Collapse
Affiliation(s)
- Qi Tan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | | | |
Collapse
|
98
|
Dormer NH, Qiu Y, Lydick AM, Allen ND, Mohan N, Berkland CJ, Detamore MS. Osteogenic differentiation of human bone marrow stromal cells in hydroxyapatite-loaded microsphere-based scaffolds. Tissue Eng Part A 2011; 18:757-67. [PMID: 21992088 DOI: 10.1089/ten.tea.2011.0176] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Calcium-based minerals have consistently been shown to stimulate osteoblastic behavior in vitro and in vivo. Thus, use of such minerals in biomaterial applications has become an effective method to enhance bone tissue engineered constructs. In the present study, for the first time, human bone marrow stromal cells (hBMSC) were osteogenically differentiated on scaffolds consisting only of hydroxyapatite (HAp)-loaded poly(D,L-lactic acid-co-glycolic acid) (PLGA) microspheres of high monodispersity. Scaffold formulations included 0, 5, 10, and 20 wt% Hap, and the hBMSC were cultured for 6 weeks. Results demonstrated suppression of some osteogenic genes during differentiation in the HAp group, but higher end-point glycosaminoglycan and collagen content in 10% and 20% HAp samples, as evidenced by biochemical tests, histology, and immunohistochemistry. After 6 weeks of culture, constructs with 0% and 5% HAp had average compressive moduli of 0.7 ± 0.2 and 1.5 ± 0.9 kPa, respectively, whereas constructs with 10% and 20% HAp had higher average moduli of 17.6 ± 4.6 and 18.9 ± 8.1 kPa, respectively. The results of this study indicate that HAp inclusion in microsphere-based scaffolds could be implemented as a physical gradient in combination with bioactive signal gradients seen in previous iterations of these microsphere-based scaffolds to enhance osteoconduction and mechanical integrity of a healing site.
Collapse
|
99
|
Montanucci P, Basta G, Pescara T, Pennoni I, Giovanni FD, Calafiore R. New Simple and Rapid Method for Purification of Mesenchymal Stem Cells from the Human Umbilical Cord Wharton Jelly. Tissue Eng Part A 2011; 17:2651-61. [DOI: 10.1089/ten.tea.2010.0587] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Pia Montanucci
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Basta
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Teresa Pescara
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Ilaria Pennoni
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Francesca Di Giovanni
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Riccardo Calafiore
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
100
|
McGuirk J, Weiss M. Promising cellular therapeutics for prevention or management of graft-versus-host disease (a review). Placenta 2011; 32 Suppl 4:S304-10. [PMID: 21658764 PMCID: PMC3760226 DOI: 10.1016/j.placenta.2011.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 04/04/2011] [Accepted: 04/17/2011] [Indexed: 12/14/2022]
Abstract
Graft-versus-host disease (GVHD) frequently occurs following allogeneic hematopoietic stem cell transplantation. The primary treatment for GVHD involves immune suppression by glucocorticoids. If patients become refractory to steroids, they have a poor prognosis. Therefore, there is a pressing need for alternative therapies to treat GVHD. Here, we review clinical data which demonstrate that a cellular therapy using mesenchymal stromal cells (MSCs) is safe and effective for GVHD. Since MSCs derived from bone marrow present certain limitations (such as time lag for expansion to clinical dose, expansion failure in vitro, painful and invasive bone marrow MSC isolation procedures), alternative sources of MSCs for cellular therapy are being sought. Here, we review data which support the notion that MSCs derived from Wharton's jelly (WJ) may be a safe and effective cellular therapy for GVHD. Many laboratories have investigated the immune properties of these discarded MSCs with an eye towards their potential use in cellular therapy. We also review data which support the notion that the licensing of MSCs (meaning the activation of MSCs by prior exposure to cytokines such as interferon-γ) may enhance their effectiveness for treatment of GVHD. In conclusion, WJCs can be collected safely and painlessly from individuals at birth, similar to the collection of cord blood, and stored cryogenically for later clinical use. Therefore, WJCs should be tested as a second generation, off-the-shelf cell therapy for the prevention or treatment of immune disorders such as GVHD.
Collapse
Affiliation(s)
- J.P. McGuirk
- University of Kansas Medical Center, Blood and Marrow Transplant Program, KS, USA
| | - M.L. Weiss
- Kansas State University, Dept of Anatomy and Physiology, Manhattan, KS 66506, USA
| |
Collapse
|