51
|
Lee HJ, Ryu JM, Jung YH, Lee KH, Kim DI, Han HJ. Glycerol-3-phosphate acyltransferase-1 upregulation by O-GlcNAcylation of Sp1 protects against hypoxia-induced mouse embryonic stem cell apoptosis via mTOR activation. Cell Death Dis 2016; 7:e2158. [PMID: 27010859 PMCID: PMC4823928 DOI: 10.1038/cddis.2015.410] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
Oxygen signaling is critical for stem cell regulation, and oxidative stress-induced stem cell apoptosis decreases the efficiency of stem cell therapy. Hypoxia activates O-linked β-N-acetyl glucosaminylation (O-GlcNAcylation) of stem cells, which contributes to regulation of cellular metabolism, as well as cell fate. Our study investigated the role of O-GlcNAcylation via glucosamine in the protection of hypoxia-induced apoptosis of mouse embryonic stem cells (mESCs). Hypoxia increased mESCs apoptosis in a time-dependent manner. Moreover, hypoxia also slightly increased the O-GlcNAc level. Glucosamine treatment further enhanced the O-GlcNAc level and prevented hypoxia-induced mESC apoptosis, which was suppressed by O-GlcNAc transferase inhibitors. In addition, hypoxia regulated several lipid metabolic enzymes, whereas glucosamine increased expression of glycerol-3-phosphate acyltransferase-1 (GPAT1), a lipid metabolic enzyme producing lysophosphatidic acid (LPA). In addition, glucosamine-increased O-GlcNAcylation of Sp1, which subsequently leads to Sp1 nuclear translocation and GPAT1 expression. Silencing of GPAT1 by gpat1 siRNA transfection reduced glucosamine-mediated anti-apoptosis in mESCs and reduced mammalian target of rapamycin (mTOR) phosphorylation. Indeed, LPA prevented mESCs from undergoing hypoxia-induced apoptosis and increased phosphorylation of mTOR and its substrates (S6K1 and 4EBP1). Moreover, mTOR inactivation by rapamycin (mTOR inhibitor) increased pro-apoptotic proteins expressions and mESC apoptosis. Furthermore, transplantation of non-targeting siRNA and glucosamine-treated mESCs increased cell survival and inhibited flap necrosis in mouse skin flap model. Conversely, silencing of GPAT1 expression reversed those glucosamine effects. In conclusion, enhancing O-GlcNAcylation of Sp1 by glucosamine stimulates GPAT1 expression, which leads to inhibition of hypoxia-induced mESC apoptosis via mTOR activation.
Collapse
Affiliation(s)
- H J Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - J M Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea
| | - Y H Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - K H Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - D I Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - H J Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
52
|
Kwon H, Paschos NK, Hu JC, Athanasiou K. Articular cartilage tissue engineering: the role of signaling molecules. Cell Mol Life Sci 2016; 73:1173-94. [PMID: 26811234 PMCID: PMC5435375 DOI: 10.1007/s00018-015-2115-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 12/10/2015] [Indexed: 02/08/2023]
Abstract
Effective early disease modifying options for osteoarthritis remain lacking. Tissue engineering approach to generate cartilage in vitro has emerged as a promising option for articular cartilage repair and regeneration. Signaling molecules and matrix modifying agents, derived from knowledge of cartilage development and homeostasis, have been used as biochemical stimuli toward cartilage tissue engineering and have led to improvements in the functionality of engineered cartilage. Clinical translation of neocartilage faces challenges, such as phenotypic instability of the engineered cartilage, poor integration, inflammation, and catabolic factors in the arthritic environment; these can all contribute to failure of implanted neocartilage. A comprehensive understanding of signaling molecules involved in osteoarthritis pathogenesis and their actions on engineered cartilage will be crucial. Thus, while it is important to continue deriving inspiration from cartilage development and homeostasis, it has become increasingly necessary to incorporate knowledge from osteoarthritis pathogenesis into cartilage tissue engineering.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Nikolaos K Paschos
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kyriacos Athanasiou
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA.
| |
Collapse
|
53
|
Synergistic effects of hypoxia and morphogenetic factors on early chondrogenic commitment of human embryonic stem cells in embryoid body culture. Stem Cell Rev Rep 2016; 11:228-41. [PMID: 25618295 DOI: 10.1007/s12015-015-9584-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Derivation of articular chondrocytes from human stem cells would advance our current understanding of chondrogenesis, and accelerate development of new stem cell therapies for cartilage repair. Chondrogenic differentiation of human embryonic stem cells (hESCs) has been studied using supplemental and cell-secreted morphogenetic factors. The use of bioreactors enabled insights into the effects of physical forces and controlled oxygen tension. In this study, we investigated the interactive effects of controlled variation of oxygen tension and chondrocyte-secreted morphogenetic factors on chondrogenic differentiation of hESCs in the embryoid body format (hESC-EB). Transient hypoxic culture (2 weeks at 5 % O2 followed by 1 week at 21 % O2) of hESC-EBs in medium conditioned with primary chondrocytes up-regulated the expression of SOX9 and suppressed pluripotent markers OCT4 and NANOG. Pellets derived from these cells showed significant up-regulation of chondrogenic genes (SOX9, COL2A1, ACAN) and enhanced production of cartilaginous matrix (collagen type II and proteoglycan) as compared to the pellets from hESC-EBs cultured under normoxic conditions. Gene expression profiles corresponded to those associated with native cartilage development, with early expression of N-cadherin (indicator of cell condensation) and late expression of aggrecan (ACAN, indicator of proteoglycan production). When implanted into highly vascularized subcutaneous area in immunocompromised mice for 4 weeks, pellets remained phenotypically stable and consisted of cartilaginous extracellular matrix (ECM), without evidence of dedifferentiation or teratoma formation. Based on these results, we propose that chondrogenesis in hESC can be synergistically enhanced by a control of oxygen tension and morphogenetic factors secreted by chondrocytes.
Collapse
|
54
|
Hammoud AA, Kirstein N, Mournetas V, Darracq A, Broc S, Blanchard C, Zeineddine D, Mortada M, Boeuf H. Murine Embryonic Stem Cell Plasticity Is Regulated through Klf5 and Maintained by Metalloproteinase MMP1 and Hypoxia. PLoS One 2016; 11:e0146281. [PMID: 26731538 PMCID: PMC4701481 DOI: 10.1371/journal.pone.0146281] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/15/2015] [Indexed: 12/12/2022] Open
Abstract
Mouse embryonic stem cells (mESCs) are expanded and maintained pluripotent in vitro in the presence of leukemia inhibitory factor (LIF), an IL6 cytokine family member which displays pleiotropic functions, depending on both cell maturity and cell type. LIF withdrawal leads to heterogeneous differentiation of mESCs with a proportion of the differentiated cells apoptosising. During LIF withdrawal, cells sequentially enter a reversible and irreversible phase of differentiation during which LIF addition induces different effects. However the regulators and effectors of LIF-mediated reprogramming are poorly understood. By employing a LIF-dependent 'plasticity' test, that we set up, we show that Klf5, but not JunB is a key LIF effector. Furthermore PI3K signaling, required for the maintenance of mESC pluripotency, has no effect on mESC plasticity while displaying a major role in committed cells by stimulating expression of the mesodermal marker Brachyury at the expense of endoderm and neuroectoderm lineage markers. We also show that the MMP1 metalloproteinase, which can replace LIF for maintenance of pluripotency, mimics LIF in the plasticity window, but less efficiently. Finally, we demonstrate that mESCs maintain plasticity and pluripotency potentials in vitro under hypoxic/physioxic growth conditions at 3% O2 despite lower levels of Pluri and Master gene expression in comparison to 20% O2.
Collapse
Affiliation(s)
- Aya Abou Hammoud
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
- Lebanese University, Beyrouth, Liban
| | - Nina Kirstein
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
| | - Virginie Mournetas
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
| | - Anais Darracq
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
| | - Sabine Broc
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
| | - Camille Blanchard
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
| | | | | | - Helene Boeuf
- Univ. Bordeaux, CIRID, UMR5164, F-33 000 Bordeaux, France
- CNRS, CIRID, UMR 5164, F-33 000 Bordeaux, France
- * E-mail:
| |
Collapse
|
55
|
Green JD, Tollemar V, Dougherty M, Yan Z, Yin L, Ye J, Collier Z, Mohammed MK, Haydon RC, Luu HH, Kang R, Lee MJ, Ho SH, He TC, Shi LL, Athiviraham A. Multifaceted signaling regulators of chondrogenesis: Implications in cartilage regeneration and tissue engineering. Genes Dis 2015; 2:307-327. [PMID: 26835506 PMCID: PMC4730920 DOI: 10.1016/j.gendis.2015.09.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/16/2015] [Indexed: 01/08/2023] Open
Abstract
Defects of articular cartilage present a unique clinical challenge due to its poor self-healing capacity and avascular nature. Current surgical treatment options do not ensure consistent regeneration of hyaline cartilage in favor of fibrous tissue. Here, we review the current understanding of the most important biological regulators of chondrogenesis and their interactions, to provide insight into potential applications for cartilage tissue engineering. These include various signaling pathways, including: fibroblast growth factors (FGFs), transforming growth factor β (TGF-β)/bone morphogenic proteins (BMPs), Wnt/β-catenin, Hedgehog, Notch, hypoxia, and angiogenic signaling pathways. Transcriptional and epigenetic regulation of chondrogenesis will also be discussed. Advances in our understanding of these signaling pathways have led to promising advances in cartilage regeneration and tissue engineering.
Collapse
Affiliation(s)
- Jordan D. Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Viktor Tollemar
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mark Dougherty
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhengjian Yan
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liangjun Yin
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zachary Collier
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maryam K. Mohammed
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin H. Ho
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
56
|
Tissue engineering of the temporomandibular joint disc: current status and future trends. Int J Artif Organs 2015; 38:55-68. [PMID: 25744198 DOI: 10.5301/ijao.5000393] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2014] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Temporomandibular joint disorders are extremely prevalent and there is no ideal treatment clinically for the moment. For severe cases, a discectomy often need to be performed, which will further result in the development of osteoarthritis. In the past thirty years, tissue engineering has provided a promising approach for the effective remedy of severe TMJ disease through the creation of viable, effective, and biological functional implants. METHODS Although TMJ disc tissue engineering is still in early stage, unremitting efforts and some achievements have been made over the past decades. In this review, a comprehensive summary of the available literature on the progress and status in tissue engineering of the TMJ disc regarding cell sources, scaffolds, biochemical and biomechanical stimuli, and other prospects relative to this field is provided. RESULTS AND CONCLUSIONS Even though research studies in this field are too few compared to other fibrocartilage (e.g., knee meniscus) and numerous, difficult tasks still exist, we believe that our ultimate goal of regenerating a biological implant whose histological, biochemical, and biomechanical properties parallel native TMJ discs for clinical therapy will be achieved in the near future.
Collapse
|
57
|
Abstract
Anchorage-dependent cells are of great interest for various biotechnological applications. (i) They represent a formidable production means of viruses for vaccination purposes at very large scales (in 1000-6000 l reactors) using microcarriers, and in the last decade many more novel viral vaccines have been developed using this production technology. (ii) With the advent of stem cells and their use/potential use in clinics for cell therapy and regenerative medicine purposes, the development of novel culture devices and technologies for adherent cells has accelerated greatly with a view to the large-scale expansion of these cells. Presently, the really scalable systems--microcarrier/microcarrier-clump cultures using stirred-tank reactors--for the expansion of stem cells are still in their infancy. Only laboratory scale reactors of maximally 2.5 l working volume have been evaluated because thorough knowledge and basic understanding of critical issues with respect to cell expansion while retaining pluripotency and differentiation potential, and the impact of the culture environment on stem cell fate, etc., are still lacking and require further studies. This article gives an overview on critical issues common to all cell culture systems for adherent cells as well as specifics for different types of stem cells in view of small- and large-scale cell expansion and production processes.
Collapse
|
58
|
Athanasiou KA, Responte DJ, Brown WE, Hu JC. Harnessing biomechanics to develop cartilage regeneration strategies. J Biomech Eng 2015; 137:020901. [PMID: 25322349 DOI: 10.1115/1.4028825] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Indexed: 12/24/2022]
Abstract
As this review was prepared specifically for the American Society of Mechanical Engineers H.R. Lissner Medal, it primarily discusses work toward cartilage regeneration performed in Dr. Kyriacos A. Athanasiou's laboratory over the past 25 years. The prevalence and severity of degeneration of articular cartilage, a tissue whose main function is largely biomechanical, have motivated the development of cartilage tissue engineering approaches informed by biomechanics. This article provides a review of important steps toward regeneration of articular cartilage with suitable biomechanical properties. As a first step, biomechanical and biochemical characterization studies at the tissue level were used to provide design criteria for engineering neotissues. Extending this work to the single cell and subcellular levels has helped to develop biochemical and mechanical stimuli for tissue engineering studies. This strong mechanobiological foundation guided studies on regenerating hyaline articular cartilage, the knee meniscus, and temporomandibular joint (TMJ) fibrocartilage. Initial tissue engineering efforts centered on developing biodegradable scaffolds for cartilage regeneration. After many years of studying scaffold-based cartilage engineering, scaffoldless approaches were developed to address deficiencies of scaffold-based systems, resulting in the self-assembling process. This process was further improved by employing exogenous stimuli, such as hydrostatic pressure, growth factors, and matrix-modifying and catabolic agents, both singly and in synergistic combination to enhance neocartilage functional properties. Due to the high cell needs for tissue engineering and the limited supply of native articular chondrocytes, costochondral cells are emerging as a suitable cell source. Looking forward, additional cell sources are investigated to render these technologies more translatable. For example, dermis isolated adult stem (DIAS) cells show potential as a source of chondrogenic cells. The challenging problem of enhanced integration of engineered cartilage with native cartilage is approached with both familiar and novel methods, such as lysyl oxidase (LOX). These diverse tissue engineering strategies all aim to build upon thorough biomechanical characterizations to produce functional neotissue that ultimately will help combat the pressing problem of cartilage degeneration. As our prior research is reviewed, we look to establish new pathways to comprehensively and effectively address the complex problems of musculoskeletal cartilage regeneration.
Collapse
|
59
|
Pimton P, Lecht S, Stabler CT, Johannes G, Schulman ES, Lelkes PI. Hypoxia enhances differentiation of mouse embryonic stem cells into definitive endoderm and distal lung cells. Stem Cells Dev 2014; 24:663-76. [PMID: 25226206 DOI: 10.1089/scd.2014.0343] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We investigated the effects of hypoxia on spontaneous (SP)- and activin A (AA)-induced definitive endoderm (DE) differentiation of mouse embryonic stem cells (mESCs) and their subsequent differentiation into distal pulmonary epithelial cells. SP differentiation for 6 days of mESCs toward endoderm at hypoxia of 1% O2, but not at 3% or 21% (normoxia), increased the expression of Sox17 and Foxa2 by 31- and 63-fold above maintenance culture, respectively. Treatment of mESCs with 20 ng/mL AA for 6 days under hypoxia further increased the expression of DE marker genes Sox17, Foxa2, and Cxcr4 by 501-, 1,483-, and 126-fold above maintenance cultures, respectively. Transient exposure to hypoxia, as short as 24 h, was sufficient to enhance AA-induced endoderm formation. The involvement of hypoxia-inducible factor (HIF)-1α and reactive oxygen species (ROS) in the AA-induced endoderm enrichment was assessed using HIF-1α(-/-) mESCs and the ROS scavenger N-acetylcysteine (NAC). Under SP conditions, HIF-1α(-/-) mESCs failed to increase the expression of endodermal marker genes but rather shifted toward ectoderm. Hypoxia induced only a marginal potentiation of AA-induced endoderm differentiation in HIF-1α(-/-) mESCs. Treatment of mESCs with AA and NAC led to a dose-dependent decrease in Sox17 and Foxa2 expression. In addition, the duration of exposure to hypoxia in the course of a recently reported lung differentiation protocol resulted in differentially enhanced expression of distal lung epithelial cell marker genes aquaporin 5 (Aqp5), surfactant protein C (Sftpc), and secretoglobin 1a1 (Scgb1a1) for alveolar epithelium type I, type II, and club cells, respectively. Our study is the first to show the effects of in vitro hypoxia on efficient formation of DE and lung lineages. We suggest that the extent of hypoxia and careful timing may be important components of in vitro differentiation bioprocesses for the differential generation of distal lung epithelial cells from pluripotent progenitors.
Collapse
Affiliation(s)
- Pimchanok Pimton
- 1 Department of Biology, School of Science, Walailak University , Nakhon Si Thammarat, Thailand
| | | | | | | | | | | |
Collapse
|
60
|
Developing functional musculoskeletal tissues through hypoxia and lysyl oxidase-induced collagen cross-linking. Proc Natl Acad Sci U S A 2014; 111:E4832-41. [PMID: 25349395 DOI: 10.1073/pnas.1414271111] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The inability to recapitulate native tissue biomechanics, especially tensile properties, hinders progress in regenerative medicine. To address this problem, strategies have focused on enhancing collagen production. However, manipulating collagen cross-links, ubiquitous throughout all tissues and conferring mechanical integrity, has been underinvestigated. A series of studies examined the effects of lysyl oxidase (LOX), the enzyme responsible for the formation of collagen cross-links. Hypoxia-induced endogenous LOX was applied in multiple musculoskeletal tissues (i.e., cartilage, meniscus, tendons, ligaments). Results of these studies showed that both native and engineered tissues are enhanced by invoking a mechanism of hypoxia-induced pyridinoline (PYR) cross-links via intermediaries like LOX. Hypoxia was shown to enhance PYR cross-linking 1.4- to 6.4-fold and, concomitantly, to increase the tensile properties of collagen-rich tissues 1.3- to 2.2-fold. Direct administration of exogenous LOX was applied in native cartilage and neocartilage generated using a scaffold-free, self-assembling process of primary chondrocytes. Exogenous LOX was found to enhance native tissue tensile properties 1.9-fold. LOX concentration- and time-dependent increases in PYR content (∼ 16-fold compared with controls) and tensile properties (approximately fivefold compared with controls) of neocartilage were also detected, resulting in properties on par with native tissue. Finally, in vivo subcutaneous implantation of LOX-treated neocartilage in nude mice promoted further maturation of the neotissue, enhancing tensile and PYR content approximately threefold and 14-fold, respectively, compared with in vitro controls. Collectively, these results provide the first report, to our knowledge, of endogenous (hypoxia-induced) and exogenous LOX applications for promoting collagen cross-linking and improving the tensile properties of a spectrum of native and engineered tissues both in vitro and in vivo.
Collapse
|
61
|
Atkinson SP, Lako M, Armstrong L. Potential for pharmacological manipulation of human embryonic stem cells. Br J Pharmacol 2014; 169:269-89. [PMID: 22515554 DOI: 10.1111/j.1476-5381.2012.01978.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The therapeutic potential of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) is vast, allowing disease modelling, drug discovery and testing and perhaps most importantly regenerative therapies. However, problems abound; techniques for cultivating self-renewing hESCs tend to give a heterogeneous population of self-renewing and partially differentiated cells and general include animal-derived products that can be cost-prohibitive for large-scale production, and effective lineage-specific differentiation protocols also still remain relatively undefined and are inefficient at producing large amounts of cells for therapeutic use. Furthermore, the mechanisms and signalling pathways that mediate pluripotency and differentiation are still to be fully appreciated. However, over the recent years, the development/discovery of a range of effective small molecule inhibitors/activators has had a huge impact in hESC biology. Large-scale screening techniques, coupled with greater knowledge of the pathways involved, have generated pharmacological agents that can boost hESC pluripotency/self-renewal and survival and has greatly increased the efficiency of various differentiation protocols, while also aiding the delineation of several important signalling pathways. Within this review, we hope to describe the current uses of small molecule inhibitors/activators in hESC biology and their potential uses in the future.
Collapse
|
62
|
Wu J, Rostami MR, Cadavid Olaya DP, Tzanakakis ES. Oxygen transport and stem cell aggregation in stirred-suspension bioreactor cultures. PLoS One 2014; 9:e102486. [PMID: 25032842 PMCID: PMC4102498 DOI: 10.1371/journal.pone.0102486] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/19/2014] [Indexed: 01/16/2023] Open
Abstract
Stirred-suspension bioreactors are a promising modality for large-scale culture of 3D aggregates of pluripotent stem cells and their progeny. Yet, cells within these clusters experience limitations in the transfer of factors and particularly O2 which is characterized by low solubility in aqueous media. Cultured stem cells under different O2 levels may exhibit significantly different proliferation, viability and differentiation potential. Here, a transient diffusion-reaction model was built encompassing the size distribution and ultrastructural characteristics of embryonic stem cell (ESC) aggregates. The model was coupled to experimental data from bioreactor and static cultures for extracting the effective diffusivity and kinetics of consumption of O2 within mouse (mESC) and human ESC (hESC) clusters. Under agitation, mESC aggregates exhibited a higher maximum consumption rate than hESC aggregates. Moreover, the reaction-diffusion model was integrated with a population balance equation (PBE) for the temporal distribution of ESC clusters changing due to aggregation and cell proliferation. Hypoxia was found to be negligible for ESCs with a smaller radius than 100 µm but became appreciable for aggregates larger than 300 µm. The integrated model not only captured the O2 profile both in the bioreactor bulk and inside ESC aggregates but also led to the calculation of the duration that fractions of cells experience a certain range of O2 concentrations. The approach described in this study can be employed for gaining a deeper understanding of the effects of O2 on the physiology of stem cells organized in 3D structures. Such frameworks can be extended to encompass the spatial and temporal availability of nutrients and differentiation factors and facilitate the design and control of relevant bioprocesses for the production of stem cell therapeutics.
Collapse
Affiliation(s)
- Jincheng Wu
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Mahboubeh Rahmati Rostami
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Diana P. Cadavid Olaya
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
63
|
Garreta E, Melo E, Navajas D, Farré R. Low oxygen tension enhances the generation of lung progenitor cells from mouse embryonic and induced pluripotent stem cells. Physiol Rep 2014; 2:2/7/e12075. [PMID: 25347858 PMCID: PMC4187564 DOI: 10.14814/phy2.12075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Whole-organ decellularization technology has emerged as a new alternative for the fabrication of bioartificial lungs. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) are potentially useful for recellularization since they can be directed to express phenotypic marker genes of lung epithelial cells. Normal pulmonary development takes place in a low oxygen environment ranging from 1 to 5%. By contrast, in vitro ESC and iPSC differentiation protocols are usually carried out at room-air oxygen tension. Here, we sought to determine the role played by oxygen tension on the derivation of Nkx2.1+ lung/thyroid progenitor cells from mouse ESC and iPSC. A step-wise differentiation protocol was used to generate Nkx2.1+ lung/thyroid progenitors under 20% and 5% oxygen tension. On day 12, gene expression analysis revealed that Nkx2.1 and Foxa2 (endodermal and early lung epithelial cell marker) were significantly upregulated at 5% oxygen tension in ESC and iPSC differentiated cultures compared to 20% oxygen conditions. In addition, quantification of Foxa2+Nkx2.1+Pax8- cells corresponding to the lung field, with exclusion of the potential thyroid fate identified by Pax8 expression, confirmed that the low physiologic oxygen tension exerted a significant positive effect on early pulmonary differentiation of ESC and iPSC. In conclusion, we found that 5% oxygen tension enhanced the derivation of lung progenitors from mouse ESC and iPSC compared to 20% room-air oxygen tension.
Collapse
Affiliation(s)
- Elena Garreta
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centre de Medicina Regenerativa de Barcelona (CMRB), Parc de Recerca Biomèdica de Barcelona (PRBB), Dr. Aiguader88 7ª Planta, Barcelona, 08003, Spain
| | - Esther Melo
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain F. Hoffmann-La Roche, AG, NORD DTABldg. 69/331, Basel, CH-4070, Switzerland
| | - Daniel Navajas
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
64
|
Hawkins KE, Sharp TV, McKay TR. The role of hypoxia in stem cell potency and differentiation. Regen Med 2014; 8:771-82. [PMID: 24147532 DOI: 10.2217/rme.13.71] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regenerative medicine relies on harnessing the capacity of stem cells to grow, divide and differentiate safely and predictably. This may be in the context of expanding stem cells in vitro or encouraging their expansion, mobilization and capacity to regenerate tissues either locally or remotely in vivo. In either case, understanding the stem cell niche is fundamental to recapitulating or manipulating conditions to enable therapy. It has become obvious that hypoxia plays a fundamental role in the maintenance of the stem cell niche. Low O2 benefits the self-renewal of human embryonic, hematopoietic, mesenchymal and neural stem cells, as well as improving the efficiency of genetic reprogramming to induced pluripotency. There is emerging evidence that harnessing or manipulating the hypoxic response can result in safer, more efficacious methodologies for regenerative medicine.
Collapse
Affiliation(s)
- Kate E Hawkins
- Division of Biomedical Sciences, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | | | | |
Collapse
|
65
|
Fynes K, Tostoes R, Ruban L, Weil B, Mason C, Veraitch FS. The differential effects of 2% oxygen preconditioning on the subsequent differentiation of mouse and human pluripotent stem cells. Stem Cells Dev 2014; 23:1910-22. [PMID: 24734982 DOI: 10.1089/scd.2013.0504] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A major challenge facing the development of effective cell therapies is the efficient differentiation of pluripotent stem cells (PSCs) into pure populations. Lowering oxygen tension to physiological levels can affect both the expansion and differentiation stages. However, to date, there are no studies investigating the knock-on effect of culturing PSCs under low oxygen conditions on subsequent lineage commitment at ambient oxygen levels. PSCs were passaged three times at 2% O2 before allowing cells to spontaneously differentiate as embryoid bodies (EBs) in high oxygen (20% O2) conditions. Maintenance of mouse PSCs in low oxygen was associated with a significant increase in the expression of early differentiation markers FGF5 and Eomes, while conversely we observed decreased expression of these genes in human PSCs. Low oxygen preconditioning primed mouse PSCs for their subsequent differentiation into mesodermal and endodermal lineages, as confirmed by increased gene expression of Eomes, Goosecoid, Brachyury, AFP, Sox17, FoxA2, and protein expression of Brachyury, Eomes, Sox17, FoxA2, relative to high oxygen cultures. The effects extended to the subsequent formation of more mature mesodermal lineages. We observed significant upregulation of cardiomyocyte marker Nkx2.5, and critically a decrease in the number of contaminant pluripotent cells after 12 days using a directed cardiomyocyte protocol. However, the impact of low oxygen preconditioning was to prime human cells for ectodermal lineage commitment during subsequent EB differentiation, with significant upregulation of Nestin and β3-tubulin. Our research demonstrates the importance of oxygen tension control during cell maintenance on the subsequent differentiation of both mouse and human PSCs, and highlights the differential effects.
Collapse
Affiliation(s)
- Kate Fynes
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London , London, United Kingdom
| | | | | | | | | | | |
Collapse
|
66
|
Kalpakci KN, Brown WE, Hu JC, Athanasiou KA. Cartilage tissue engineering using dermis isolated adult stem cells: the use of hypoxia during expansion versus chondrogenic differentiation. PLoS One 2014; 9:e98570. [PMID: 24867063 PMCID: PMC4035316 DOI: 10.1371/journal.pone.0098570] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/04/2014] [Indexed: 11/25/2022] Open
Abstract
Dermis isolated adult stem (DIAS) cells, a subpopulation of dermis cells capable of chondrogenic differentiation in the presence of cartilage extracellular matrix, are a promising source of autologous cells for tissue engineering. Hypoxia, through known mechanisms, has profound effects on in vitro chondrogenesis of mesenchymal stem cells and could be used to improve the expansion and differentiation processes for DIAS cells. The objective of this study was to build upon the mechanistic knowledge of hypoxia and translate it to tissue engineering applications to enhance chondrogenic differentiation of DIAS cells through exposure to hypoxic conditions (5% O2) during expansion and/or differentiation. DIAS cells were isolated and expanded in hypoxic (5% O2) or normoxic (20% O2) conditions, then differentiated for 2 weeks in micromass culture on chondroitin sulfate-coated surfaces in both environments. Monolayer cells were examined for proliferation rate and colony forming efficiency. Micromasses were assessed for cellular, biochemical, and histological properties. Differentiation in hypoxic conditions following normoxic expansion increased per cell production of collagen type II 2.3 fold and glycosaminoglycans 1.2 fold relative to continuous normoxic culture (p<0.0001). Groups expanded in hypoxia produced 51% more collagen and 23% more GAGs than those expanded in normoxia (p<0.0001). Hypoxia also limited cell proliferation in monolayer and in 3D culture. Collectively, these data show hypoxic differentiation following normoxic expansion significantly enhances chondrogenic differentiation of DIAS cells, improving the potential utility of these cells for cartilage engineering.
Collapse
Affiliation(s)
- Kerem N. Kalpakci
- Medtronic Spine & Biologics, Memphis, Tennessee, United States of America
| | - Wendy E. Brown
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
| | - Jerry C. Hu
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
| | - Kyriacos A. Athanasiou
- UC Davis, Department of Biomedical Engineering, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
67
|
Paschos NK, Brown WE, Eswaramoorthy R, Hu JC, Athanasiou KA. Advances in tissue engineering through stem cell-based co-culture. J Tissue Eng Regen Med 2014; 9:488-503. [PMID: 24493315 DOI: 10.1002/term.1870] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/19/2013] [Accepted: 01/03/2014] [Indexed: 12/13/2022]
Abstract
Stem cells are the future in tissue engineering and regeneration. In a co-culture, stem cells not only provide a target cell source with multipotent differentiation capacity, but can also act as assisting cells that promote tissue homeostasis, metabolism, growth and repair. Their incorporation into co-culture systems seems to be important in the creation of complex tissues or organs. In this review, critical aspects of stem cell use in co-culture systems are discussed. Direct and indirect co-culture methodologies used in tissue engineering are described, along with various characteristics of cellular interactions in these systems. Direct cell-cell contact, cell-extracellular matrix interaction and signalling via soluble factors are presented. The advantages of stem cell co-culture strategies and their applications in tissue engineering and regenerative medicine are portrayed through specific examples for several tissues, including orthopaedic soft tissues, bone, heart, vasculature, lung, kidney, liver and nerve. A concise review of the progress and the lessons learned are provided, with a focus on recent developments and their implications. It is hoped that knowledge developed from one tissue can be translated to other tissues. Finally, we address challenges in tissue engineering and regenerative medicine that can potentially be overcome via employing strategies for stem cell co-culture use.
Collapse
Affiliation(s)
- Nikolaos K Paschos
- Department of Biomedical Engineering and Orthopedic Surgery, University of California at Davis, CA, 95616, USA
| | | | | | | | | |
Collapse
|
68
|
Brown PT, Handorf AM, Jeon WB, Li WJ. Stem cell-based tissue engineering approaches for musculoskeletal regeneration. Curr Pharm Des 2013; 19:3429-45. [PMID: 23432679 DOI: 10.2174/13816128113199990350] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/10/2013] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine and tissue engineering is an ever evolving field that holds promise in treating numerous musculoskeletal diseases and injuries. An important impetus in the development of the field was the discovery and implementation of stem cells. The utilization of mesenchymal stem cells, and later embryonic and induced pluripotent stem cells, opens new arenas for tissue engineering and presents the potential of developing stem cell-based therapies for disease treatment. Multipotent and pluripotent stem cells can produce various lineage tissues, and allow for derivation of a tissue that may be comprised of multiple cell types. As the field grows, the combination of biomaterial scaffolds and bioreactors provides methods to create an environment for stem cells that better represent their microenvironment for new tissue formation. As technologies for the fabrication of biomaterial scaffolds advance, the ability of scaffolds to modulate stem cell behavior advances as well. The composition of scaffolds could be of natural or synthetic materials and could be tailored to enhance cell self-renewal and/or direct cell fates. In addition to biomaterial scaffolds, studies of tissue development and cellular microenvironments have determined other factors, such as growth factors and oxygen tension, that are crucial to the regulation of stem cell activity. The overarching goal of stem cell-based tissue engineering research is to precisely control differentiation of stem cells in culture. In this article, we review current developments in tissue engineering, focusing on several stem cell sources, induction factors including growth factors, oxygen tension, biomaterials, and mechanical stimulation, and the internal and external regulatory mechanisms that govern proliferation and differentiation.
Collapse
Affiliation(s)
- Patrick T Brown
- Wisconsin Institutes of Medical Research, 1111 Highland Ave., Madison, WI 53705, USA
| | | | | | | |
Collapse
|
69
|
Hypoxia enhances chondrogenic differentiation of human adipose tissue-derived stromal cells in scaffold-free and scaffold systems. Cell Tissue Res 2013; 355:89-102. [PMID: 24178804 DOI: 10.1007/s00441-013-1732-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 09/05/2013] [Indexed: 02/05/2023]
Abstract
Human adipose-derived stromal cells (hASCs) possess the potential for chondrogenic differentiation. Recent studies imply that this differentiation process may be enhanced by culturing the cells in low oxygen tension in combination with three-dimensional (3D) scaffolds. We report the evaluation of the chondrogenic potential of hASC pellets in 5 and 21% O2 and as cell-scaffold constructs using a collagen I/III scaffold with chemical induction using TGF-β3. hASCs from four human donors were cultured both in a micromass pellet system and in 3D collagen I/III scaffolds in either 5 or 21% O2. Chondrogenesis was evaluated by quantitative gene expression analysis of aggrecan, SOX9, collagen I, II and X and histological evaluation with H&E and toluidine blue staining. Induced pellets cultured in 5% O2 showed increased peripheral cellularity and matrix deposition compared with 21% O2. Induced pellets cultured in 5% O2 had increased control-adjusted gene expression of aggrecan, SOX9 and collagen I and decreased collagen X compared with 21% O2 cultures. Induced pellets had higher gene expression of aggrecan, SOX9, collagen I, II and X and increased ratios of collagen II/I and collagen II/X compared with controls. As for pellets, scaffold cultures showed cellularity and matrix deposition organized in a zonal manner as a function of the oxygen tension, with a cartilage-like morphology and matrix deposition peripherally in the 5% O2 group and a more centrally located matrix in the 21% O2 group. There were no differences in histology and gene expressions between pellet and scaffold cultures. Five percent O2 in combination with chondrogenic culture medium stimulated chondrogenic differentiation of hASCs in vitro. We observed similar patterns of differentiation and matrix disposition in pellet and scaffold cultures.
Collapse
|
70
|
Cheng A, Hardingham TE, Kimber SJ. Generating cartilage repair from pluripotent stem cells. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:257-66. [PMID: 23957872 DOI: 10.1089/ten.teb.2012.0757] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The treatment of degeneration and injury of articular cartilage has been very challenging for scientists and surgeons. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble. Autologous chondrocyte implantation for cartilage defects has achieved good results, but the limited resources and complexity of the procedure have hindered wider application. Stem cells form an alternative to chondrocytes as a source of chondrogenic cells due to their ability to proliferate extensively while retaining the potential for differentiation. Adult stem cells such as mesenchymal stem cells have been differentiated into chondrocytes, but the limitations in their proliferative ability and the heterogeneous cell population hinder their adoption as a prime alternative source for generating chondrocytes. Human embryonic stem cells (hESCs) are attractive as candidates for cell replacement therapy because of their unlimited self-renewal and ability for differentiation into mesodermal derivatives as well as other lineages. In this review, we focus on current protocols for chondrogenic differentiation of ESCs, in particular the chemically defined culture system developed in our lab that could potentially be adapted for clinical application.
Collapse
Affiliation(s)
- Aixin Cheng
- 1 North West Embryonic Stem Cell Centre, Faculty of Life Science, University of Manchester , Manchester, United Kingdom
| | | | | |
Collapse
|
71
|
Effects of severe hypoxia on bone marrow mesenchymal stem cells differentiation potential. Stem Cells Int 2013; 2013:232896. [PMID: 24082888 PMCID: PMC3777136 DOI: 10.1155/2013/232896] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 06/27/2013] [Accepted: 06/30/2013] [Indexed: 02/06/2023] Open
Abstract
Background. The interests in mesenchymal stem cells (MSCs) and their application in cell therapy have resulted in a better understanding of the basic biology of these cells. Recently hypoxia has been indicated as crucial for complete chondrogenesis. We aimed at analyzing bone marrow MSCs (BM-MSCs) differentiation capacity under normoxic and severe hypoxic culture conditions. Methods. MSCs were characterized by flow cytometry and differentiated towards adipocytes, osteoblasts, and chondrocytes under normoxic or severe hypoxic conditions. The differentiations were confirmed comparing each treated point with a control point made of cells grown in DMEM and fetal bovine serum (FBS). Results. BM-MSCs from the donors displayed only few phenotypical differences in surface antigens expressions. Analyzing marker genes expression levels of the treated cells compared to their control point for each lineage showed a good differentiation in normoxic conditions and the absence of this differentiation capacity in severe hypoxic cultures. Conclusions. In our experimental conditions, severe hypoxia affects the in vitro differentiation potential of BM-MSCs. Adipogenic, osteogenic, and chondrogenic differentiations are absent in severe hypoxic conditions. Our work underlines that severe hypoxia slows cell differentiation by means of molecular mechanisms since a decrease in the expression of adipocyte-, osteoblast-, and chondrocyte-specific genes was observed.
Collapse
|
72
|
Knuth CA, Clark ME, Meeson AP, Khan SK, Dowen DJ, Deehan DJ, Oldershaw RA. Low Oxygen Tension is Critical for the Culture of Human Mesenchymal Stem Cells with Strong Osteogenic Potential from Haemarthrosis Fluid. Stem Cell Rev Rep 2013; 9:599-608. [DOI: 10.1007/s12015-013-9446-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
73
|
Yodmuang S, Gadjanski I, Chao PHG, Vunjak-Novakovic G. Transient hypoxia improves matrix properties in tissue engineered cartilage. J Orthop Res 2013. [PMID: 23203946 PMCID: PMC4136653 DOI: 10.1002/jor.22275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Adult articular cartilage is a hypoxic tissue, with oxygen tension ranging from <10% at the cartilage surface to <1% in the deepest layers. In addition to spatial gradients, cartilage development is also associated with temporal changes in oxygen tension. However, a vast majority of cartilage tissue engineering protocols involves cultivation of chondrocytes or their progenitors under ambient oxygen concentration (21% O(2)), that is, significantly above physiological levels in either developing or adult cartilage. Our study was designed to test the hypothesis that transient hypoxia followed by normoxic conditions results in improved quality of engineered cartilaginous ECM. To this end, we systematically compared the effects of normoxia (21% O(2) for 28 days), hypoxia (5% O(2) for 28 days) and transient hypoxia--reoxygenation (5% O(2) for 7 days and 21% O(2) for 21 days) on the matrix composition and expression of the chondrogenic genes in cartilage constructs engineered in vitro. We demonstrated that reoxygenation had the most effect on the expression of cartilaginous genes including COL2A1, ACAN, and SOX9 and increased tissue concentrations of amounts of glycosaminoglycans and type II collagen. The equilibrium Young's moduli of tissues grown under transient hypoxia (510.01 ± 28.15 kPa) and under normoxic conditions (417.60 ± 68.46 kPa) were significantly higher than those measured under hypoxic conditions (279.61 ± 20.52 kPa). These data suggest that the cultivation protocols utilizing transient hypoxia with reoxygenation have high potential for efficient cartilage tissue engineering, but need further optimization in order to achieve higher mechanical functionality of engineered constructs.
Collapse
Affiliation(s)
- Supansa Yodmuang
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Ivana Gadjanski
- Department of Biomedical Engineering, Columbia University, New York, New York
- R&D Center for Bioengineering, Metropolitan University Belgrade, Prvoslava Stojanovica 6, Kragujevac 34000, Serbia
| | - Pen-hsiu Grace Chao
- Institute of Biomedical Engineering, School of Engineering and School of Medicine, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
74
|
Spaapen F, van den Akker GGH, Caron MMJ, Prickaerts P, Rofel C, Dahlmans VEH, Surtel DAM, Paulis Y, Schweizer F, Welting TJM, Eijssen LM, Voncken JW. The immediate early gene product EGR1 and polycomb group proteins interact in epigenetic programming during chondrogenesis. PLoS One 2013; 8:e58083. [PMID: 23483971 PMCID: PMC3590300 DOI: 10.1371/journal.pone.0058083] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 01/30/2013] [Indexed: 12/18/2022] Open
Abstract
Initiation of and progression through chondrogenesis is driven by changes in the cellular microenvironment. At the onset of chondrogenesis, resting mesenchymal stem cells are mobilized in vivo and a complex, step-wise chondrogenic differentiation program is initiated. Differentiation requires coordinated transcriptomic reprogramming and increased progenitor proliferation; both processes require chromatin remodeling. The nature of early molecular responses that relay differentiation signals to chromatin is poorly understood. We here show that immediate early genes are rapidly and transiently induced in response to differentiation stimuli in vitro. Functional ablation of the immediate early factor EGR1 severely deregulates expression of key chondrogenic control genes at the onset of differentiation. In addition, differentiating cells accumulate DNA damage, activate a DNA damage response and undergo a cell cycle arrest and prevent differentiation associated hyper-proliferation. Failed differentiation in the absence of EGR1 affects global acetylation and terminates in overall histone hypermethylation. We report novel molecular connections between EGR1 and Polycomb Group function: Polycomb associated histone H3 lysine27 trimethylation (H3K27me3) blocks chromatin access of EGR1. In addition, EGR1 ablation results in abnormal Ezh2 and Bmi1 expression. Consistent with this functional interaction, we identify a number of co-regulated targets genes in a chondrogenic gene network. We here describe an important role for EGR1 in early chondrogenic epigenetic programming to accommodate early gene-environment interactions in chondrogenesis.
Collapse
Affiliation(s)
- Frank Spaapen
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Guus G. H. van den Akker
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marjolein M. J. Caron
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Peggy Prickaerts
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Celine Rofel
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Vivian E. H. Dahlmans
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Don A. M. Surtel
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Yvette Paulis
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Finja Schweizer
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Tim J. M. Welting
- Department of Orthopaedic Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Lars M. Eijssen
- Department of Bioinformatics – BiGCaT, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Willem Voncken
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
- * E-mail:
| |
Collapse
|
75
|
Wu CL, Diekman BO, Jain D, Guilak F. Diet-induced obesity alters the differentiation potential of stem cells isolated from bone marrow, adipose tissue and infrapatellar fat pad: the effects of free fatty acids. Int J Obes (Lond) 2012; 37:1079-87. [PMID: 23164698 PMCID: PMC3582830 DOI: 10.1038/ijo.2012.171] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/30/2012] [Accepted: 09/09/2012] [Indexed: 02/06/2023]
Abstract
Introduction Obesity is a major risk factor for several musculoskeletal conditions that are characterized by an imbalance of tissue remodeling. Adult stem cells are closely associated with the remodeling and potential repair of several mesodermally derived tissues such as fat, bone, and cartilage. We hypothesized that obesity would alter the frequency, proliferation, multipotency, and immunophenotype of adult stem cells from a variety of tissues. Materials and Methods Bone marrow-derived mesenchymal stem cells (MSCs), subcutaneous adipose-derived stem cells (sqASCs), and infrapatellar fat pad-derived stem cells (IFP cells) were isolated from lean and high-fat diet induced obese mice, and their cellular properties were examined. To test the hypothesis that changes in stem cell properties were due to the increased systemic levels of free fatty acids (FFAs), we further investigated the effects of FFAs on lean stem cells in vitro. Results Obese mice showed a trend toward increased prevalence of MSCs and sqASCs in the stromal tissues. While no significant differences in cell proliferation were observed in vitro, the differentiation potential of all types of stem cells was altered by obesity. MSCs from obese mice demonstrated decreased adipogenic, osteogenic, and chondrogenic potential. Obese sqASCs and IFP cells showed increased adipogenic and osteogenic differentiation, but decreased chondrogenic ability. Obese MSCs also showed decreased CD105 and increased PDGFRα expression, consistent with decreased chondrogenic potential. FFA treatment of lean stem cells significantly altered their multipotency but did not completely recapitulate the properties of obese stem cells. Conclusions These findings support the hypothesis that obesity alters the properties of adult stem cells in a manner that depends on the cell source. These effects may be regulated in part by increased levels of FFAs, but may involve other obesity-associated cytokines. These findings contribute to our understanding of mesenchymal tissue remodeling with obesity, as well as the development of autologous stem cell therapies for obese patients.
Collapse
Affiliation(s)
- C-L Wu
- Departments of Orthopaedic Surgery and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | | | | | | |
Collapse
|
76
|
Human stem cells and articular cartilage regeneration. Cells 2012; 1:994-1009. [PMID: 24710539 PMCID: PMC3901135 DOI: 10.3390/cells1040994] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/15/2012] [Accepted: 10/24/2012] [Indexed: 01/12/2023] Open
Abstract
The regeneration of articular cartilage damaged due to trauma and posttraumatic osteoarthritis is an unmet medical need. Current approaches to regeneration and tissue engineering of articular cartilage include the use of chondrocytes, stem cells, scaffolds and signals, including morphogens and growth factors. Stem cells, as a source of cells for articular cartilage regeneration, are a critical factor for articular cartilage regeneration. This is because articular cartilage tissue has a low cell turnover and does not heal spontaneously. Adult stem cells have been isolated from various tissues, such as bone marrow, adipose, synovial tissue, muscle and periosteum. Signals of the transforming growth factor beta superfamily play critical roles in chondrogenesis. However, adult stem cells derived from various tissues tend to differ in their chondrogenic potential. Pluripotent stem cells have unlimited proliferative capacity compared to adult stem cells. Chondrogenesis from embryonic stem (ES) cells has been studied for more than a decade. However, establishment of ES cells requires embryos and leads to ethical issues for clinical applications. Induced pluripotent stem (iPS) cells are generated by cellular reprogramming of adult cells by transcription factors. Although iPS cells have chondrogenic potential, optimization, generation and differentiation toward articular chondrocytes are currently under intense investigation.
Collapse
|
77
|
Gregory-Evans CY, Wallace VA, Gregory-Evans K. Gene networks: dissecting pathways in retinal development and disease. Prog Retin Eye Res 2012; 33:40-66. [PMID: 23128416 DOI: 10.1016/j.preteyeres.2012.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 01/21/2023]
Abstract
During retinal neurogenesis, diverse cellular subtypes originate from multipotent neural progenitors in a spatiotemporal order leading to a highly specialized laminar structure combined with a distinct mosaic architecture. This is driven by the combinatorial action of transcription factors and signaling molecules which specify cell fate and differentiation. The emerging approach of gene network analysis has allowed a better understanding of the functional relationships between genes expressed in the developing retina. For instance, these gene networks have identified transcriptional hubs that have revealed potential targets and pathways for the development of therapeutic options for retinal diseases. Much of the current knowledge has been informed by targeted gene deletion experiments and gain-of-functional analysis. In this review we will provide an update on retinal development gene networks and address the wider implications for future disease therapeutics.
Collapse
Affiliation(s)
- Cheryl Y Gregory-Evans
- Department of Ophthalmology, University of British Columbia, Vancouver, BC V5Z 3N9, Canada.
| | | | | |
Collapse
|
78
|
Lee SW, Jeong HK, Lee JY, Yang J, Lee EJ, Kim SY, Youn SW, Lee J, Kim WJ, Kim KW, Lim JM, Park JW, Park YB, Kim HS. Hypoxic priming of mESCs accelerates vascular-lineage differentiation through HIF1-mediated inverse regulation of Oct4 and VEGF. EMBO Mol Med 2012; 4:924-38. [PMID: 22821840 PMCID: PMC3491825 DOI: 10.1002/emmm.201101107] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 05/15/2012] [Accepted: 05/18/2012] [Indexed: 12/21/2022] Open
Abstract
Hypoxic microenvironment plays an important role in determining stem cell fates. However, it is controversial to which direction between self-renewal and differentiation the hypoxia drives the stem cells. Here, we investigated whether a short exposure to hypoxia (termed ‘hypoxic-priming’) efficiently directed and promoted mouse embryonic stem cells (mESCs) to differentiate into vascular-lineage. During spontaneous differentiation of embryoid bodies (EBs), hypoxic region was observed inside EB spheroids even under normoxic conditions. Indeed, hypoxia-primed EBs more efficiently differentiated into cells of vascular-lineage, than normoxic EBs did. We found that hypoxia suppressed Oct4 expression via direct binding of HIF-1 to reverse hypoxia-responsive elements (rHREs) in the Oct4 promoter. Furthermore, vascular endothelial growth factor (VEGF) was highly upregulated in hypoxia-primed EBs, which differentiated towards endothelial cells in the absence of exogenous VEGF. Interestingly, this differentiation was abolished by the HIF-1 or VEGF blocking. In vivo transplantation of hypoxia-primed EBs into mice ischemic limb elicited enhanced vessel differentiation. Collectively, our findings identify that hypoxia enhanced ESC differentiation by HIF-1-mediated inverse regulation of Oct4 and VEGF, which is a novel pathway to promote vascular-lineage differentiation.
Collapse
Affiliation(s)
- Sae-Won Lee
- Department of Internal Medicine, Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Duval E, Baugé C, Andriamanalijaona R, Bénateau H, Leclercq S, Dutoit S, Poulain L, Galéra P, Boumédiene K. Molecular mechanism of hypoxia-induced chondrogenesis and its application in in vivo cartilage tissue engineering. Biomaterials 2012; 33:6042-51. [PMID: 22677190 DOI: 10.1016/j.biomaterials.2012.04.061] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/30/2012] [Indexed: 11/29/2022]
Abstract
Cartilage engineering is one of the most challenging issue in regenerative medicine, due to its limited self-ability to repair. Here, we assessed engineering of cartilage tissue starting from human bone marrow (hBM) stem cells under hypoxic environment and delineated the mechanism whereby chondrogenesis could be conducted without addition of exogenous growth factors. hBM stem cells were cultured in alginate beads and chondrogenesis was monitored by chondrocyte phenotypic markers. Activities and roles of Sox and HIF-1α transcription factors were investigated with complementary approaches of gain and loss of function and provided evidences that HIF-1α is essential for hypoxic induction of chondrogenesis. Thereafter, hBM cells and human articular chondrocytes (HAC) underwent chondrogenesis by 3D and hypoxic culture for 7 days or by ectopic expression of HIF-1α. After subcutaneous implantation of 3 weeks into athymic mice, tissue analysis showed that hypoxia or HIF-1α overexpression is effective and sufficient to induce chondrocyte phenotype in hBM cells, without use of exogenous growth factors. Therefore, this study brings interesting data for a simple and affordable system in biotechnology of cartilage engineering.
Collapse
Affiliation(s)
- Elise Duval
- Université de Caen Basse Normandie, MILPAT, Caen 14032, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Bae D, Mondragon-Teran P, Hernandez D, Ruban L, Mason C, Bhattacharya SS, Veraitch FS. Hypoxia Enhances the Generation of Retinal Progenitor Cells from Human Induced Pluripotent and Embryonic Stem Cells. Stem Cells Dev 2012; 21:1344-55. [DOI: 10.1089/scd.2011.0225] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Daekyeong Bae
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Paul Mondragon-Teran
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Diana Hernandez
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Ludmila Ruban
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Chris Mason
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Shomi S. Bhattacharya
- CABIMER, Isla de la Cartuja, Sevilla, Spain
- UCL-Institute of Ophthalmology, London, United Kingdom
| | - Farlan S. Veraitch
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
81
|
Ko EC, Fujihara Y, Ogasawara T, Asawa Y, Nishizawa S, Nagata S, Takato T, Hoshi K. BMP-2 Embedded Atelocollagen Scaffold for Tissue-Engineered Cartilage Cultured in the Medium Containing Insulin and Triiodothyronine—A New Protocol for Three-Dimensional In Vitro Culture of Human Chondrocytes. Tissue Eng Part C Methods 2012; 18:374-86. [DOI: 10.1089/ten.tec.2011.0217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Edward Chengchuan Ko
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- School of Dentistry, Collge of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Oral and Maxillofacial Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yuko Fujihara
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
| | - Toru Ogasawara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiyo Asawa
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
| | - Satoru Nishizawa
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
| | - Satoru Nagata
- Nagata Microtia and Reconstructive Plastic Surgery Clinic, Saitama, Japan
| | - Tsuyoshi Takato
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuto Hoshi
- Departments of Cartilage and Bone Regeneration (Fujisoft), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
82
|
Huey DJ, Athanasiou KA. Tension-compression loading with chemical stimulation results in additive increases to functional properties of anatomic meniscal constructs. PLoS One 2011; 6:e27857. [PMID: 22114714 PMCID: PMC3218070 DOI: 10.1371/journal.pone.0027857] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/26/2011] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to improve the functional properties of anatomically-shaped meniscus constructs through simultaneous tension and compression mechanical stimulation in conjunction with chemical stimulation. METHODS Scaffoldless meniscal constructs were subjected to simultaneous tension and compressive stimulation and chemical stimulation. The temporal aspect of mechanical loading was studied by employing two separate five day stimulation periods. Chemical stimulation consisted of the application of a catabolic GAG-depleting enzyme, chondroitinase ABC (C-ABC), and an anabolic growth factor, TGF-β1. Mechanical and chemical stimulation combinations were studied through a full-factorial experimental design and assessed for histological, biochemical, and biomechanical properties following 4 wks of culture. RESULTS Mechanical loading applied from days 10-14 resulted in significant increases in compressive, tensile, and biochemical properties of meniscal constructs. When mechanical and chemical stimuli were combined significant additive increases in collagen per wet weight (4-fold), compressive instantaneous (3-fold) and relaxation (2-fold) moduli, and tensile moduli in the circumferential (4-fold) and radial (6-fold) directions were obtained. CONCLUSIONS This study demonstrates that a stimulation regimen of simultaneous tension and compression mechanical stimulation, C-ABC, and TGF-β1 is able to create anatomic meniscus constructs replicating the compressive mechanical properties, and collagen and GAG content of native tissue. In addition, this study significantly advances meniscus tissue engineering by being the first to apply simultaneous tension and compression mechanical stimulation and observe enhancement of tensile and compressive properties following mechanical stimulation.
Collapse
Affiliation(s)
- Daniel J. Huey
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| |
Collapse
|
83
|
Potential of human embryonic stem cells in cartilage tissue engineering and regenerative medicine. Stem Cell Rev Rep 2011; 7:544-59. [PMID: 21188652 DOI: 10.1007/s12015-010-9222-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The current surgical intervention of using autologous chondrocyte implantation (ACI) for cartilage repair is associated with several problems such as donor site morbidity, de-differentiation upon expansion and fibrocartilage repair following transplantation. This has led to exploration of the use of stem cells as a model for chondrogenic differentiation as well as a potential source of chondrogenic cells for cartilage tissue engineering and repair. Embryonic stem cells (ESCs) are advantageous, due to their unlimited self-renewal and pluripotency, thus representing an immortal cell source that could potentially provide an unlimited supply of chondrogenic cells for both cell and tissue-based therapies and replacements. This review aims to present an overview of emerging trends of using ESCs in cartilage tissue engineering and regenerative medicine. In particular, we will be focusing on ESCs as a promising cell source for cartilage regeneration, the various strategies and approaches employed in chondrogenic differentiation and tissue engineering, the associated outcomes from animal studies, and the challenges that need to be overcome before clinical application is possible.
Collapse
|
84
|
Boström KI, Rajamannan NM, Towler DA. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ Res 2011; 109:564-77. [PMID: 21852555 DOI: 10.1161/circresaha.110.234278] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular calcification increasingly afflicts our aging, dysmetabolic population. Once considered only a passive process of dead and dying cells, vascular calcification has now emerged as a highly regulated form of biomineralization organized by collagenous and elastin extracellular matrices. During skeletal bone formation, paracrine epithelial-mesenchymal and endothelial-mesenchymal interactions control osteochondrocytic differentiation of multipotent mesenchymal progenitor cells. These paracrine osteogenic signals, mediated by potent morphogens of the bone morphogenetic protein and wingless-type MMTV integration site family member (Wnt) superfamilies, are also active in the programming of arterial osteoprogenitor cells during vascular and valve calcification. Inflammatory cytokines, reactive oxygen species, and oxylipids-increased in the clinical settings of atherosclerosis, diabetes, and uremia that promote arteriosclerotic calcification-elicit the ectopic vascular activation of osteogenic morphogens. Specific extracellular and intracellular inhibitors of bone morphogenetic protein-Wnt signaling have been identified as contributing to the regulation of osteogenic mineralization during development and disease. These inhibitory pathways and their regulators afford the development of novel therapeutic strategies to prevent and treat valve and vascular sclerosis.
Collapse
Affiliation(s)
- Kristina I Boström
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 10833 LeConte Ave, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
85
|
Punwar S, Khan WS. Mesenchymal stem cells and articular cartilage repair: clinical studies and future direction. Open Orthop J 2011. [PMID: 21886696 DOI: 10.2174/187432500110501029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cartilage is frequently injured but shows little capacity for repair. Current treatment options include the use of procedures that stimulate repair through the stimulation of subchondral bone marrow and result in the formation of fibrocartilage. There is considerable interest in the use of cell-based treatment strategies and there are limited studies describing the use of mesenchymal stem cells for cartilage repair with promising early results. This paper reviews the current treatment strategies for articular cartilage, describes use of mesenchymal stem cells for articular cartilage repair along with the results of clinical studies, and describes the future direction that these strategies are likely to take.
Collapse
Affiliation(s)
- Shahid Punwar
- Department of Trauma and Orthopaedics, Frenchay Hospital, North Bristol NHS Trust, Bristol, BS16 1LE, UK
| | | |
Collapse
|
86
|
Punwar S, Khan WS. Mesenchymal stem cells and articular cartilage repair: clinical studies and future direction. Open Orthop J 2011; 5 Suppl 2:296-301. [PMID: 21886696 PMCID: PMC3149861 DOI: 10.2174/1874325001105010296] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 02/24/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022] Open
Abstract
Cartilage is frequently injured but shows little capacity for repair. Current treatment options include the use of procedures that stimulate repair through the stimulation of subchondral bone marrow and result in the formation of fibrocartilage. There is considerable interest in the use of cell-based treatment strategies and there are limited studies describing the use of mesenchymal stem cells for cartilage repair with promising early results. This paper reviews the current treatment strategies for articular cartilage, describes use of mesenchymal stem cells for articular cartilage repair along with the results of clinical studies, and describes the future direction that these strategies are likely to take.
Collapse
Affiliation(s)
- Shahid Punwar
- Department of Trauma and Orthopaedics, Frenchay Hospital, North Bristol NHS Trust, Bristol, BS16 1LE, UK
| | | |
Collapse
|
87
|
Effects of oxygen and culture system on in vitro propagation and redifferentiation of osteoarthritic human articular chondrocytes. Cell Tissue Res 2011; 347:649-63. [PMID: 21638206 DOI: 10.1007/s00441-011-1193-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 05/12/2011] [Indexed: 02/06/2023]
Abstract
Regenerative medicine-based approaches for the repair of damaged cartilage rely on the ability to propagate cells while promoting their chondrogenic potential. Thus, conditions for cell expansion should be optimized through careful environmental control. Appropriate oxygen tension and cell expansion substrates and controllable bioreactor systems are probably critical for expansion and subsequent tissue formation during chondrogenic differentiation. We therefore evaluated the effects of oxygen and microcarrier culture on the expansion and subsequent differentiation of human osteoarthritic chondrocytes. Freshly isolated chondrocytes were expanded on tissue culture plastic or CultiSpher-G microcarriers under hypoxic or normoxic conditions (5% or 20% oxygen partial pressure, respectively) followed by cell phenotype analysis with flow cytometry. Cells were redifferentiated in micromass pellet cultures over 4 weeks, under either hypoxia or normoxia. Chondrocytes cultured on tissue culture plastic proliferated faster, expressed higher levels of cell surface markers CD44 and CD105 and demonstrated stronger staining for proteoglycans and collagen type II in pellet cultures compared with microcarrier-cultivated cells. Pellet wet weight, glycosaminoglycan content and expression of chondrogenic genes were significantly increased in cells differentiated under hypoxia. Hypoxia-inducible factor-3α mRNA was up-regulated in these cultures in response to low oxygen tension. These data confirm the beneficial influence of reduced oxygen on ex vivo chondrogenesis. However, hypoxia during cell expansion and microcarrier bioreactor culture does not enhance intrinsic chondrogenic potential. Further improvements in cell culture conditions are therefore required before chondrocytes from osteoarthritic and aged patients can become a useful cell source for cartilage regeneration.
Collapse
|
88
|
Foldager CB, Nielsen AB, Munir S, Ulrich-Vinther M, Søballe K, Bünger C, Lind M. Combined 3D and hypoxic culture improves cartilage-specific gene expression in human chondrocytes. Acta Orthop 2011; 82:234-40. [PMID: 21434761 PMCID: PMC3235297 DOI: 10.3109/17453674.2011.566135] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE In vitro expansion of autologous chondrocytes is an essential part of many clinically used cartilage repair treatments. Native chondrocytes reside in a 3-dimensional (3D) network and are exposed to low levels of oxygen. We compared monolayer culture to combined 3D and hypoxic culture using quantitative gene expression analysis. METHODS Cartilage biopsies were collected from the intercondylar groove in the distal femur from 12 patients with healthy cartilage. Cells were used for either monolayer or scaffold culture. The scaffolds were clinically available MPEG-PLGA scaffolds (ASEED). After harvesting of cells for baseline investigation, the remainder was divided into 3 groups for incubation in conditions of normoxia (21% oxygen), hypoxia (5% oxygen), or severe hypoxia (1% oxygen). RNA extractions were performed 1, 2, and 6 days after the baseline time point, respectively. Quantitative RT-PCR was performed using assays for RNA encoding collagen types 1 and 2, aggrecan, sox9, ankyrin repeat domain-37, and glyceraldehyde-3-phosphate dehydrogenase relative to 2 hypoxia-stable housekeeping genes. RESULTS Sox9, aggrecan, and collagen type 2 RNA expression increased with reduced oxygen. On day 6, the expression of collagen type 2 and aggrecan RNA was higher in 3D culture than in monolayer culture. INTERPRETATION Our findings suggest that there was a combined positive effect of 3D culture and hypoxia on cartilage-specific gene expression. The positive effects of 3D culture alone were not detected until day 6, suggesting that seeding of chondrocytes onto a scaffold for matrix-assisted chondrocyte implantation should be performed earlier than 2 days before implantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Martin Lind
- Sports Trauma Clinic, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
89
|
Ko EC, Fujihara Y, Ogasawara T, Asawa Y, Nishizawa S, Nagata S, Takato T, Hoshi K. Administration of the insulin into the scaffold atelocollagen for tissue-engineered cartilage. J Biomed Mater Res A 2011; 97:186-92. [DOI: 10.1002/jbm.a.33046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 10/27/2010] [Accepted: 11/22/2010] [Indexed: 11/08/2022]
|
90
|
Cicione C, Díaz-Prado S, Muiños-López E, Hermida-Gómez T, Blanco FJ. Molecular profile and cellular characterization of human bone marrow mesenchymal stem cells: donor influence on chondrogenesis. Differentiation 2011; 80:155-65. [PMID: 20619527 DOI: 10.1016/j.diff.2010.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/27/2010] [Accepted: 06/04/2010] [Indexed: 11/27/2022]
Abstract
BACKGROUND The use of autologous or allogenic stem cells has recently been suggested as an alternative therapeutic approach for treatment of cartilage defects. Bone marrow mesenchymal stem cells (BM-MSCs) are well-characterized multipotent cells that can differentiate into different cell types. Understanding the potential of these cells and the molecular mechanisms underlying their differentiation should lead to innovative protocols for clinical applications. The aim of this study was to evaluate the usefulness of surface antigen selection of BM-MSCs and to understand the mechanisms underlying their differentiation. METHODS MSCs were isolated from BM stroma and expanded. CD105+ subpopulation was isolated using a magnetic separator. We compared culture-expanded selected cells with non-selected cells. We analyzed the phenotypic profiles, the expression of the stem cell marker genes Nanog, Oct3/4, and Sox2 and the multi-lineage differentiation potential (adipogenic, osteogenic, and chondrogenic). The multi-lineage differentiation was confirmed using histochemistry, immunohistochemistry and/or real-time polymerase chain reaction (qPCR) techniques. RESULTS The selected and non-selected cells displayed similar phenotypes and multi-lineage differentiation potentials. Analyzing each cell source individually, we could divide the six donors into two groups: one with a high percentage of CD29 (β1-integrin) expression (HL); one with a low percentage of CD29 (LL). These two groups had different chondrogenic capacities and different expression levels of the stem cell marker genes. CONCLUSIONS This study showed that phenotypic profiles of donors were related to the chondrogenic potential of human BM-MSCs. The chondrogenic potential of donors was related to CD29 expression levels. The high expression of CD29 antigen seemed necessary for chondrogenic differentiation. Further investigation into the mechanisms responsible for these differences in BM-MSCs chondrogenesis is therefore warranted. Understanding the mechanisms for these differences will contribute to improved clinical use of autologous human BM-MSCs for articular cartilage repair.
Collapse
Affiliation(s)
- Claudia Cicione
- CIBER-BBN-Cellular Therapy Area, Hospital Universitario A Coruña, C/As Xubias S/N, 15.006-A Coruña, Spain
| | | | | | | | | |
Collapse
|
91
|
Sheehy EJ, Buckley CT, Kelly DJ. Chondrocytes and bone marrow-derived mesenchymal stem cells undergoing chondrogenesis in agarose hydrogels of solid and channelled architectures respond differentially to dynamic culture conditions. J Tissue Eng Regen Med 2011; 5:747-58. [DOI: 10.1002/term.385] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 10/13/2010] [Indexed: 12/18/2022]
|
92
|
Chen HC, Sytwu HK, Chang JL, Wang HW, Chen HK, Kang BH, Liu DW, Chen CH, Chao TT, Wang CH. Hypoxia enhances the stemness markers of cochlear stem/progenitor cells and expands sphere formation through activation of hypoxia-inducible factor-1 alpha. Hear Res 2010; 275:43-52. [PMID: 21147209 DOI: 10.1016/j.heares.2010.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 11/22/2010] [Accepted: 12/01/2010] [Indexed: 10/18/2022]
Abstract
Unlike neural stem cells that maintain populations in the adult brains of both rodents and humans, cochlear stem cells appear to diminish in number after birth and may become quiescent in adult mammalian cochleae. Hypoxia has been observed to promote an undifferentiated cell state in various stem cell populations; however, little is known about such an effect on cochlear stem/progenitor cells (SPCs). The aims of this study were to assess the effect of hypoxia on cochlear SPCs and to examine the impact of hypoxia-inducible factor-1 alpha (Hif-1a) on regulating such an effect. Our data demonstrate that hypoxic culturing for 24 h significantly increased sphere formation and viability of cochlear SPCs compared with those cultured under normoxic conditions. Concurrent with these proliferation promotion effects are changes in the expression of multiple stemness and cell-cycle quiescent associated gene targets, including Abcg2, nestin, p27(Kip1)and Vegf. Knockdown of Hif-1a expression by small-interfering RNA inhibited hypoxia-induced cochlear SPC expansion and resulted in downregulation of Vegf, Abcg2, and nestin and upregulation of p27(Kip1) gene expression. These results suggest that Hif-1a plays an important role in the stimulation of the proliferation of cochlear SPCs, which confers a great benefit of expanding cochlear SPCs via hypoxic conditions.
Collapse
Affiliation(s)
- Hsin-Chien Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Teramura T, Onodera Y, Mihara T, Hosoi Y, Hamanishi C, Fukuda K. Induction of mesenchymal progenitor cells with chondrogenic property from mouse-induced pluripotent stem cells. Cell Reprogram 2010; 12:249-61. [PMID: 20698767 DOI: 10.1089/cell.2009.0086] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite recent cell-engineering advances, treatment and repair of cartilage remains challenging. Although stem cell transplantation therapy using mesenchymal stem cells (MSCs) is considered a prominent strategy, the major problem of limited proliferative capacity of autologous cells has been unsolved. Recently, an induced pluripotent stem (iPS) cell line was suggested as an alternative way to cure various human diseases due to their potential proliferating infinitely while possessing the capacity to form all types of cells. However, the method to induce lineage-restricted differentiation has not been well examined or established. Here, we suggest a simple method to induce mesenchymal progenitors possessing chondrogenic property from mouse iPS cells. The MSC-like cells produced in our study expressed some MSC markers, and could also differentiate to osteoblast and adipocyte. The present study demonstrates the property of iPS cells as an alternative candidate for treatment of articular disorders, and suggests an effective approach for preparing chondrocyte from iPS cells.
Collapse
Affiliation(s)
- Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kinki University School of Medicine, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
94
|
Yang Y, Kusano K, Frei H, Rossi F, Brunette DM, Putnins EE. Microtopographical regulation of adult bone marrow progenitor cells chondrogenic and osteogenic gene and protein expressions. J Biomed Mater Res A 2010; 95:294-304. [PMID: 20607868 DOI: 10.1002/jbm.a.32838] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Microtopographic features affect diverse cell behaviors. Adult bone marrow progenitor cells (AMPCs) constitute a multipotent heterogeneous population. We hypothesized that microtopographies could direct AMPCs lineage-specific differentiation. AMPCs isolated from Sprague-Dawley rats were CD45 depleted, expanded, and plated at 10(5) cells/cm2 on epoxy-microfabricated: (1) 60-microm-deep grooves with 95-microm pitch (D60P95), (2) 55-microm-wide and 10-microm-deep squares (W55D10), (3) 30-microm-deep grooves with 45-microm pitch (D30P45), (4) 17-microm-wide and 10-microm-deep pillars (W17D10), and (5) smooth control. AMPCs were cultured using expansion, chondrogenesis, or osteogenesis supporting media. Cell cultures were examined by scanning electron microscopy, qRT-PCR, and immunostaining at 2, 9, 16, and 23 days after plating. Expressions of osteogenesis-related genes, such as Runx-2, alkaline phosphatase, osteopontin, osteocalcin, and parathyroid hormone-related protein receptor (PTHr), and chondrogenesis-associated genes, such as Sox-9, type II collagen, and aggrecan, were determined. In expansion medium, W55D10 induced a transient increase of Sox9 expression. Compared with smooth surfaces, type II collagen mRNA and protein expressions in chondrogenic medium were significantly upregulated on W55D10 by day 23. In contrast, osteocalcin and PTHr expressions were significantly increased on D30P45 in osteogenic medium. We have demonstrated that W55D10 and D30P45 enhanced AMPCs chondrogenic and osteogenic terminal differentiation with appropriate culture conditions.
Collapse
Affiliation(s)
- Y Yang
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, 2199 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | | | | | | | | | | |
Collapse
|
95
|
Buckley CT, Vinardell T, Kelly DJ. Oxygen tension differentially regulates the functional properties of cartilaginous tissues engineered from infrapatellar fat pad derived MSCs and articular chondrocytes. Osteoarthritis Cartilage 2010; 18:1345-54. [PMID: 20650328 DOI: 10.1016/j.joca.2010.07.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 07/07/2010] [Accepted: 07/14/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND For current tissue engineering or regenerative medicine strategies, chondrocyte (CC)- or mesenchymal stem cell (MSC)-seeded constructs are typically cultured in normoxic conditions (20% oxygen). However, within the knee joint capsule a lower oxygen tension exists. OBJECTIVE The objective of this study was to investigate how CCs and infrapatellar fad pad derived MSCs will respond to a low oxygen (5%) environment in 3D agarose culture. Our hypothesis was that culture in a low oxygen environment (5%) will enhance the functional properties of cartilaginous tissues engineered using both cell sources. EXPERIMENTAL DESIGN Cell-encapsulated agarose hydrogel constructs (seeded with CCs or infrapatellar fat pad (IFP) derived MSCs) were prepared and cultured in a chemically defined serum-free medium in the presence (CCs and MSCs) or absence (CCs only) of transforming growth factor-beta3 (TGF-β3) in normoxic (20%) or low oxygen (5%) conditions for 42 days. Constructs were assessed at days 0, 21 and 42 in terms of mechanical properties, biochemical content and histologically. RESULTS Low oxygen tension (5%) was observed to promote extracellular matrix (ECM) production by CCs cultured in the absence of TGF-β3, but was inhibitory in the presence of TGF-β3. In contrast, a low oxygen tension enhanced chondrogenesis of IFP constructs in the presence of TGF-β3, leading to superior mechanical functionality compared to CCs cultured in identical conditions. CONCLUSIONS Extrapolating the results of this study to the in vivo setting, it would appear that joint fat pad derived MSCs may possess a superior potential to generate a functional repair tissue in low oxygen tensions. However, in the context of in vitro cartilage tissue engineering, CCs maintained in normoxic conditions in the presence of TGF-β3 generate the most mechanically functional tissue.
Collapse
Affiliation(s)
- C T Buckley
- Trinity Centre for Bioengineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | | | | |
Collapse
|
96
|
Jiang YZ, Zhang SF, Qi YY, Wang LL, Ouyang HW. Cell transplantation for articular cartilage defects: principles of past, present, and future practice. Cell Transplant 2010; 20:593-607. [PMID: 20887665 DOI: 10.3727/096368910x532738] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
As articular cartilage has very limited self-repair capability, the repair and regeneration of damaged cartilage is a major challenge. This review aims to outline the past, present, and future of cell therapies for articular cartilage defect repair. Autologous chondrocyte implantation (ACI) has been used clinically for more than 20 years, and the short, medium, and long-term clinical outcomes of three generation of ACI are extensively overviewed. Also, strategies of clinical outcome evaluation, ACI limitations, and the comparison of ACI clinical outcomes with those of other surgical techniques are discussed. Moreover, mesenchymal stem cells and pluripotent stem cells for cartilage regeneration in vitro, in vivo, and in a few clinical studies are reviewed. This review not only comprehensively analyzes the ACI clinical data but also considers the findings from state-of-the-art stem cell research on cartilage repair from bench and bedside. The conclusion provides clues for the future development of strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Yang Zi Jiang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | |
Collapse
|
97
|
Jukes JM, van Blitterswijk CA, de Boer J. Skeletal tissue engineering using embryonic stem cells. J Tissue Eng Regen Med 2010; 4:165-80. [PMID: 19967745 DOI: 10.1002/term.234] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Various cell types have been investigated as candidate cell sources for cartilage and bone tissue engineering. In this review, we focused on chondrogenic and osteogenic differentiation of mouse and human embryonic stem cells (ESCs) and their potential in cartilage and bone tissue engineering. A decade ago, mouse ESCs were first used as a model to study cartilage and bone development and essential genes, factors and conditions for chondrogenesis and osteogenesis were unravelled. This knowledge, combined with data from the differentiation of adult stem cells, led to successful chondrogenic and osteogenic differentiation of mouse ESCs and later also human ESCs. Next, researchers focused on the use of ESCs for skeletal tissue engineering. Cartilage and bone tissue was formed in vivo using ESCs. However, the amount, homogeneity and stability of the cartilage and bone formed were still insufficient for clinical application. The current protocols require improvement not only in differentiation efficiency but also in ESC-specific hurdles, such as tumourigenicity and immunorejection. In addition, some of the general tissue engineering challenges, such as cell seeding and nutrient limitation in larger constructs, will also apply for ESCs. In conclusion, there are still many challenges, but there is potential for ESCs in skeletal tissue engineering.
Collapse
Affiliation(s)
- Jojanneke M Jukes
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | | | | |
Collapse
|
98
|
Prado-Lopez S, Conesa A, Armiñán A, Martínez-Losa M, Escobedo-Lucea C, Gandia C, Tarazona S, Melguizo D, Blesa D, Montaner D, Sanz-González S, Sepúlveda P, Götz S, O'Connor JE, Moreno R, Dopazo J, Burks DJ, Stojkovic M. Hypoxia promotes efficient differentiation of human embryonic stem cells to functional endothelium. Stem Cells 2010; 28:407-18. [PMID: 20049902 DOI: 10.1002/stem.295] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Early development of mammalian embryos occurs in an environment of relative hypoxia. Nevertheless, human embryonic stem cells (hESC), which are derived from the inner cell mass of blastocyst, are routinely cultured under the same atmospheric conditions (21% O(2)) as somatic cells. We hypothesized that O(2) levels modulate gene expression and differentiation potential of hESC, and thus, we performed gene profiling of hESC maintained under normoxic or hypoxic (1% or 5% O(2)) conditions. Our analysis revealed that hypoxia downregulates expression of pluripotency markers in hESC but increases significantly the expression of genes associated with angio- and vasculogenesis including vascular endothelial growth factor and angiopoitein-like proteins. Consequently, we were able to efficiently differentiate hESC to functional endothelial cells (EC) by varying O(2) levels; after 24 hours at 5% O(2), more than 50% of cells were CD34+. Transplantation of resulting endothelial-like cells improved both systolic function and fractional shortening in a rodent model of myocardial infarction. Moreover, analysis of the infarcted zone revealed that transplanted EC reduced the area of fibrous scar tissue by 50%. Thus, use of hypoxic conditions to specify the endothelial lineage suggests a novel strategy for cellular therapies aimed at repair of damaged vasculature in pathologies such as cerebral ischemia and myocardial infarction.
Collapse
Affiliation(s)
- Sonia Prado-Lopez
- Cellular Reprogramming Laboratory, Avenida del Autopista del Saler 16, 46013 Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Cartilage repair using hyaluronan hydrogel-encapsulated human embryonic stem cell-derived chondrogenic cells. Biomaterials 2010; 31:6968-80. [PMID: 20619789 DOI: 10.1016/j.biomaterials.2010.05.064] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Accepted: 05/25/2010] [Indexed: 11/23/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to offer a virtually unlimited source of chondrogenic cells for use in cartilage repair and regeneration. We have recently shown that expandable chondrogenic cells can be derived from hESCs under selective growth factor-responsive conditions. In this study, we explore the potential of these hESC-derived chondrogenic cells to produce an extracellular matrix (ECM)-enriched cartilaginous tissue construct when cultured in hyaluronic acid (HA)-based hydrogel, and further investigated the long-term reparative ability of the resulting hESC-derived chondrogenic cell-engineered cartilage (HCCEC) in an osteochondral defect model. We hypothesized that HCCEC can provide a functional template capable of undergoing orderly remodeling during the repair of critical-sized osteochondral defects (1.5 mm in diameter, 1 mm depth into the subchondral bone) in a rat model. In the process of repair, we observed an orderly spatial-temporal remodeling of HCCEC over 12 weeks into osteochondral tissue, with characteristic architectural features including a hyaline-like neocartilage layer with good surface regularity and complete integration with the adjacent host cartilage and a regenerated subchondral bone. By 12 weeks, the HCCEC-regenerated osteochondral tissue resembled closely that of age-matched unoperated native control, while only fibrous tissue filled in the control defects which left empty or treated with hydrogel alone. Here we demonstrate that transplanted hESC-derived chondrogenic cells maintain long-term viability with no evidence of tumorigenicity, providing a safe, highly-efficient and practical strategy of applying hESCs for cartilage tissue engineering.
Collapse
|
100
|
Gunja NJ, Athanasiou KA. Additive and synergistic effects of bFGF and hypoxia on leporine meniscus cell-seeded PLLA scaffolds. J Tissue Eng Regen Med 2010; 4:115-22. [PMID: 19937913 PMCID: PMC3553794 DOI: 10.1002/term.221] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Injuries to avascular regions of menisci do not heal and result in significant discomfort to patients. Current treatments, such as partial meniscectomy, alleviate these symptoms in the short term but lead to premature osteoarthritis as a result of compromised stability and changes in knee biomechanics. Thus, tissue engineering of the meniscus may provide an alternative treatment modality to overcome this problem. In this experiment, a scaffold-based tissue-engineering approach was utilized to regenerate the meniscus. Meniscus cells were cultured on poly-L-lactic acid scaffolds in normoxic (approximately 21% oxygen) or hypoxic (approximately 2% oxygen) conditions in the presence or absence of the growth factor, basic fibroblast growth factor (bFGF). At t = 4 weeks, histological sections of constructs showed presence of collagen and glycosaminoglycan (GAG) in all groups. Immunohistochemical staining showed the presence of collagen I in all groups and collagen II in groups cultured under hypoxic conditions. bFGF in the culture medium significantly increased cell number/construct by 25%, regardless of culture conditions. For GAG/construct, synergistic increases were observed in constructs cultured in hypoxic conditions and bFGF (two-fold) when compared to constructs cultured in normoxic conditions. Compressive tests showed synergistic increases in the relaxation modulus and coefficient of viscosity and additive increases in the instantaneous modulus for constructs cultured under hypoxic conditions and bFGF, when compared to constructs cultured under normoxic conditions. Overall, these results demonstrate that bFGF and hypoxia can significantly enhance the ability of meniscus cells to produce GAGs and improve the compressive properties of tissue-engineered meniscus constructs in vitro.
Collapse
Affiliation(s)
- Najmuddin J Gunja
- Department of Bioengineering, Rice University, Houston, TX 77251, USA
| | | |
Collapse
|