251
|
Wobma HM, Liu D, Vunjak-Novakovic G. Paracrine Effects of Mesenchymal Stromal Cells Cultured in Three-Dimensional Settings on Tissue Repair. ACS Biomater Sci Eng 2017; 4:1162-1175. [PMID: 33418654 DOI: 10.1021/acsbiomaterials.7b00005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising cell source for promoting tissue repair, due to their ability to release growth, angiogenic, and immunomodulatory factors. However, when injected as a suspension, these cells suffer from poor survival and localization, and suboptimal release of paracrine factors. While there have been attempts to overcome these limitations by modifying MSCs themselves, a more versatile solution is to grow them in three dimensions, as aggregates or embedded into biomaterials. Here we review the mechanisms by which 3D culture can influence the regenerative capacity of undifferentiated MSCs, focusing on recent examples from the literature. We further discuss how knowledge of these mechanisms can lead to strategic design of MSC therapies that overcome some of the challenges to their effective translation.
Collapse
|
252
|
Egger D, Schwedhelm I, Hansmann J, Kasper C. Hypoxic Three-Dimensional Scaffold-Free Aggregate Cultivation of Mesenchymal Stem Cells in a Stirred Tank Reactor. Bioengineering (Basel) 2017; 4:bioengineering4020047. [PMID: 28952526 PMCID: PMC5590473 DOI: 10.3390/bioengineering4020047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/19/2017] [Accepted: 05/21/2017] [Indexed: 01/10/2023] Open
Abstract
Extensive expansion of mesenchymal stem cells (MSCs) for cell-based therapies remains challenging since long-term cultivation and excessive passaging in two-dimensional conditions result in a loss of essential stem cell properties. Indeed, low survival rate of cells, alteration of surface marker profiles, and reduced differentiation capacity are observed after in vitro expansion and reduce therapeutic success in clinical studies. Remarkably, cultivation of MSCs in three-dimensional aggregates preserve stem cell properties. Hence, the large scale formation and cultivation of MSC aggregates is highly desirable. Besides other effects, MSCs cultivated under hypoxic conditions are known to display increased proliferation and genetic stability. Therefore, in this study we demonstrate cultivation of adipose derived human MSC aggregates in a stirred tank reactor under hypoxic conditions. Although aggregates were exposed to comparatively high average shear stress of 0.2 Pa as estimated by computational fluid dynamics, MSCs displayed a viability of 78-86% and maintained their surface marker profile and differentiation potential after cultivation. We postulate that cultivation of 3D MSC aggregates in stirred tank reactors is valuable for large-scale production of MSCs or their secreted compounds after further optimization of cultivation parameters.
Collapse
Affiliation(s)
- Dominik Egger
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| | - Ivo Schwedhelm
- Translational Center, University Hospital Wuerzburg, Roentgenring 11, 97070 Wuerzburg, Germany.
| | - Jan Hansmann
- Translational Center, University Hospital Wuerzburg, Roentgenring 11, 97070 Wuerzburg, Germany.
| | - Cornelia Kasper
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
253
|
He D, Wang RX, Mao JP, Xiao B, Chen DF, Tian W. Three-dimensional spheroid culture promotes the stemness maintenance of cranial stem cells by activating PI3K/AKT and suppressing NF-κB pathways. Biochem Biophys Res Commun 2017; 488:528-533. [PMID: 28522297 DOI: 10.1016/j.bbrc.2017.05.080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/14/2017] [Indexed: 12/11/2022]
Abstract
Multipotent stem cells are one of the most powerful tools available for the bone regeneration. However, owing to various limitations, including a lack of tissue-specific stem cell identification, reconstruction of large cranial bone defects remains challenging. In the current study, we isolated a population of Sca-1+CD105+CD140a+ stem cells from adult mouse calvarium and cultured them as three-dimensional spheroids. Although these cells shared similar surface antigens when grown in either monolayers or spheroids, the cranial stem cells grown in spheroids possessed enhanced multipotency and proliferation capacity. In addition, the cranial stem cells in spheroids were found to express high levels of the self-renewal transcription factors Nanog, Oct-4, and Sox-2. Mechanistically, we found that three-dimensional spheroid culture suppressed NF-κB pathways, but activated the PI3K/AKT pathway in cranial stem cells. More importantly, activation of NF-κB pathways or specific inhibition of the PI3K/AKT pathway partially impaired spheroid formation and suppressed expression of self-renewal transcription factors. In summary, these findings reveal a novel effect of spheroid culture in promoting the maintenance of cranial stem cell stemness and indicate that NF-κB and PI3K/AKT pathways might be involved in the stemness maintenance.
Collapse
Affiliation(s)
- Da He
- Department of Spine Surgery, Bei Jing JiShuiTan Hospital, Beijing, China
| | - Ren-Xian Wang
- Department of Bone Tissue Engineering, Bei Jing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Jian-Ping Mao
- Department of Spine Surgery, Bei Jing JiShuiTan Hospital, Beijing, China
| | - Bin Xiao
- Department of Spine Surgery, Bei Jing JiShuiTan Hospital, Beijing, China
| | - Da-Fu Chen
- Department of Bone Tissue Engineering, Bei Jing Research Institute of Traumatology and Orthopaedics, Beijing, China.
| | - Wei Tian
- Department of Spine Surgery, Bei Jing JiShuiTan Hospital, Beijing, China.
| |
Collapse
|
254
|
Petrenko Y, Syková E, Kubinová Š. The therapeutic potential of three-dimensional multipotent mesenchymal stromal cell spheroids. Stem Cell Res Ther 2017; 8:94. [PMID: 28446248 PMCID: PMC5406927 DOI: 10.1186/s13287-017-0558-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficiency of clinical trials involving transplantation of multipotent mesenchymal stromal cells (MSCs) is often insufficient due to harsh conditions present within the target tissue including hypoxia, low nutrient supply as well as inflammatory reactions. This indicates the necessity for optimization of cell-based therapy approaches which might include either modification of the cell manufacturing process or specific cell pretreatment procedures prior to transplantation. Recent reports confirm evidence that the aggregation of MSCs into three-dimensional (3D) multicellular spheroids results in enhancement of the overall therapeutic potential of cells, by improving the anti-inflammatory and angiogenic properties, stemness and survival of MSCs after transplantation. Such an MSCs spheroid generation approach may open new opportunities for the enlargement of MSCs applications in clinical research and therapy. However, the unification and optimization of 3D spheroid generation techniques, including the selection of appropriate clinical-grade culture conditions and methods for their large-scale production, are still of great importance. The current review addresses questions regarding therapeutic-associated properties of 3D multicellular MSCs spheroids in vitro and during preclinical animal studies, with special attention to the possibilities of translating these research achievements toward further clinical manufacturing and applications.
Collapse
Affiliation(s)
- Yuriy Petrenko
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine AS CR v. v. i, Vídeňská 1083, 14220, Prague 4-Krč, Czech Republic.
| | - Eva Syková
- Department of Neuroscience, Charles University, Second Faculty of Medicine, V Uvalu 84, 15006, Prague, Czech Republic
| | - Šárka Kubinová
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine AS CR v. v. i, Vídeňská 1083, 14220, Prague 4-Krč, Czech Republic
| |
Collapse
|
255
|
Ylostalo JH, Bazhanov N, Mohammadipoor A, Bartosh TJ. Production and Administration of Therapeutic Mesenchymal Stem/Stromal Cell (MSC) Spheroids Primed in 3-D Cultures Under Xeno-free Conditions. J Vis Exp 2017:55126. [PMID: 28362380 PMCID: PMC5409342 DOI: 10.3791/55126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) hold great promise in bioengineering and regenerative medicine. MSCs can be isolated from multiple adult tissues via their strong adherence to tissue culture plastic and then further expanded in vitro, most commonly using fetal bovine serum (FBS). Since FBS can cause MSCs to become immunogenic, its presence in MSC cultures limits both clinical and experimental applications of the cells. Therefore, studies employing chemically defined xeno-free (XF) media for MSC cultures are extremely valuable. Many beneficial effects of MSCs have been attributed to their ability to regulate inflammation and immunity, mainly through secretion of immunomodulatory factors such as tumor necrosis factor-stimulated gene 6 (TSG6) and prostaglandin E2 (PGE2). However, MSCs require activation to produce these factors and since the effect of MSCs is often transient, great interest has emerged to discover ways of pre-activating the cells prior to their use, thus eliminating the lag time for activation in vivo. Here we present protocols to efficiently activate or prime MSCs in three-dimensional (3D) cultures under chemically defined XF conditions and to administer these pre-activated MSCs in vivo. Specifically, we first describe methods to generate spherical MSC micro-tissues or 'spheroids' in hanging drops using XF medium and demonstrate how the spheres and conditioned medium (CM) can be harvested for various applications. Second, we describe gene expression screens and in vitro functional assays to rapidly assess the level of MSC activation in spheroids, emphasizing the anti-inflammatory and anti-cancer potential of the cells. Third, we describe a novel method to inject intact MSC spheroids into the mouse peritoneal cavity for in vivo efficacy testing. Overall, the protocols herein overcome major challenges of obtaining pre-activated MSCs under chemically defined XF conditions and provide a flexible system to administer MSC spheroids for therapies.
Collapse
Affiliation(s)
| | | | - Arezoo Mohammadipoor
- Multi-Organ Support Technology Task Area, U.S. Army Institute of Surgical Research
| | | |
Collapse
|
256
|
Zhou Y, Chen H, Li H, Wu Y. 3D culture increases pluripotent gene expression in mesenchymal stem cells through relaxation of cytoskeleton tension. J Cell Mol Med 2017; 21:1073-1084. [PMID: 28276635 PMCID: PMC5431137 DOI: 10.1111/jcmm.12946] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 07/04/2016] [Indexed: 02/06/2023] Open
Abstract
Three‐dimensional (3D) culture has been shown to improve pluripotent gene expression in mesenchymal stem cells (MSCs), but the underlining mechanisms were poorly understood. Here, we found that the relaxation of cytoskeleton tension of MSCs in 3D culture was critically associated with the expressional up‐regulation of Nanog. Cultured in spheroids, MSCs showed decreased integrin‐based cell–matrix adhesion but increased cadherin‐based cell–cell interaction. Different from that in 2D culture, where MSCs exhibited branched and multiple‐directed F‐actin stress bundles at the cell edge and strengthened stress fibres transversing the cell body, MSCs cultured in spheroids showed compact cell body, relaxed cytoskeleton tension with very thin cortical actin filament outlining the cell, and increased expression of Nanog along with reduced levels of Suv39h1 (H3K9 methyltransferase) and H3K9me3. Notably, pharmaceutical inhibition of actin polymerization with cytochalasin D or silencing Suv39h1 expression with siRNA in 2D‐cultured MSCs elevated the expression of Nanog via H3K9 demethylation. Thus, our data suggest that 3D culture increases the expression of Nanog through the relaxation of actin cytoskeleton, which mediates reduced Suv39h1 and H3K9me3 levels.
Collapse
Affiliation(s)
- Ying Zhou
- School of Life Sciences, Tsinghua University, Beijing, China.,The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Haiyan Chen
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Beijing, China
| | - Hong Li
- Department of General Surgery, Qingdao Municipal Hospital Qingdao, Qingdao, China
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Beijing, China
| |
Collapse
|
257
|
Biodegradable poly-ε-caprolactone microcarriers for efficient production of human mesenchymal stromal cells and secreted cytokines in batch and fed-batch bioreactors. Cytotherapy 2017; 19:419-432. [DOI: 10.1016/j.jcyt.2016.11.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/26/2016] [Accepted: 11/11/2016] [Indexed: 01/01/2023]
|
258
|
Tsai AC, Liu Y, Yuan X, Chella R, Ma T. Aggregation kinetics of human mesenchymal stem cells under wave motion. Biotechnol J 2017; 12. [PMID: 27996210 DOI: 10.1002/biot.201600448] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/01/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are primary candidates in cell therapy and regenerative medicine but preserving their therapeutic potency following culture expansion is a significant challenge. hMSCs can spontaneously assemble into three-dimensional (3D) aggregates that enhance their regenerative properties. The present study investigated the impact of hydrodynamics conditions on hMSC aggregation kinetics under controlled rocking motion. While various laboratory methods have been developed for hMSC aggregate production, the rocking platform provides gentle mixing and can be scaled up using large bags as in wave motion bioreactors. The results show that the hMSC aggregation is mediated by cell adhesion molecules and that aggregate size distribution is influenced by seeding density, culture time, and hydrodynamic conditions. The analysis of fluid shear stress by COMSOL indicated that aggregate size distribution is inversely correlated with shear stress and that the rocking angle had a more pronounced effect on aggregate size distribution than the rocking speed due to its impact on shear stress. hMSC aggregates obtained from the bioreactor exhibit increased stemness, migratory properties, and expression of angiogenic factors. The results demonstrate the potential of the rocking platform to produce hMSC aggregates with controlled size distribution for therapeutic application.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Yijun Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Ravindran Chella
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
259
|
Composite Bioscaffolds Incorporating Decellularized ECM as a Cell-Instructive Component Within Hydrogels as In Vitro Models and Cell Delivery Systems. Methods Mol Biol 2017; 1577:183-208. [PMID: 28493212 DOI: 10.1007/7651_2017_36] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decellularized tissues represent promising biomaterials, which harness the innate capacity of the tissue-specific extracellular matrix (ECM) to direct cell functions including stem cell proliferation and lineage-specific differentiation. However, bioscaffolds derived exclusively from decellularized ECM offer limited versatility in terms of tuning biomechanical properties, as well as cell-cell and cell-ECM interactions that are important mediators of the cellular response. As an alternative approach, in the current chapter we describe methods for incorporating cryo-milled decellularized tissues as a cell-instructive component within a hydrogel carrier designed to crosslink under mild conditions. This composite strategy can enable in situ cell encapsulation with high cell viability, allowing efficient seeding with a homogeneous distribution of cells and ECM. Detailed protocols are provided for the effective decellularization of human adipose tissue and porcine auricular cartilage, as well as the cryo-milling process used to generate the ECM particles. Further, we describe methods for synthesizing methacrylated chondroitin sulphate (MCS) and for performing UV-initiated and thermally induced crosslinking to form hydrogel carriers for adipose and cartilage regeneration. The hydrogel composites offer great flexibility, and the hydrogel phase, ECM source, particle size, cell type(s) and seeding density can be tuned to promote the desired cellular response. Overall, these systems represent promising platforms for the development of tissue-specific 3-D in vitro cell culture models and in vivo cell delivery systems.
Collapse
|
260
|
Shearier E, Xing Q, Qian Z, Zhao F. Physiologically Low Oxygen Enhances Biomolecule Production and Stemness of Mesenchymal Stem Cell Spheroids. Tissue Eng Part C Methods 2016; 22:360-9. [PMID: 26830500 DOI: 10.1089/ten.tec.2015.0465] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Multicellular human mesenchymal stem cell (hMSC) spheroids have been demonstrated to be valuable in a variety of applications, including cartilage regeneration, wound healing, and neoangiogenesis. Physiological relevant low oxygen culture can significantly improve in vitro hMSC expansion by preventing cell differentiation. We hypothesize that hypoxia-cultured hMSC spheroids can better maintain the regenerative properties of hMSCs. In this study, hMSC spheroids were fabricated using hanging drop method and cultured under 2% O2 and 20% O2 for up to 96 h. Spheroid diameter and viability were examined, as well as extracellular matrix (ECM) components and growth factor levels between the two oxygen tensions at different time points. Stemness was measured among the spheroid culture conditions and compared to two-dimensional cell cultures. Spheroid viability and structural integrity were studied using different needle gauges to ensure no damage would occur when implemented in vivo. Spheroid attachment and integration within a tissue substitute were also demonstrated. The results showed that a three-dimensional hMSC spheroid cultured at low oxygen conditions can enhance the production of ECM proteins and growth factors, while maintaining the spheroids' stemness and ability to be injected, attached, and potentially be integrated within a tissue.
Collapse
Affiliation(s)
- Emily Shearier
- Department of Biomedical Engineering, Michigan Technological University , Houghton, Michigan
| | - Qi Xing
- Department of Biomedical Engineering, Michigan Technological University , Houghton, Michigan
| | - Zichen Qian
- Department of Biomedical Engineering, Michigan Technological University , Houghton, Michigan
| | - Feng Zhao
- Department of Biomedical Engineering, Michigan Technological University , Houghton, Michigan
| |
Collapse
|
261
|
Sisakhtnezhad S, Alimoradi E, Akrami H. External factors influencing mesenchymal stem cell fate in vitro. Eur J Cell Biol 2016; 96:13-33. [PMID: 27988106 DOI: 10.1016/j.ejcb.2016.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have extensive potentials, which make them attractive candidates for the developmental biology, drug discovery and regenerative medicine. However, the use of MSCs is limited by their scarceness in tissues and in culture conditions. They also exhibit various degrees of potency which subsequently influencing their applications. Nowadays, questions remain about how self-renewal and differentiation of MSCs can be controlled in vitro and in vivo, how they will behave and migrate to the right place and how they modulate the immune system. Therefore, identification of factors and culture conditions to affect the fate and function of MSCs may be effective to enhance their applications in clinical situations. Studies have indicated that the fate of MSCs in culture is influenced by various external factors, including the specific cell source, donor age, plating density, passage number and plastic surface quality. Some other factors such as cell culture media and their supplementary factors, O2 concentration, mechano-/electro-stimuli and three-dimensional scaffolds are also shown to be influential. This review addresses the current state of MSC research for describing and discussing the findings about external factors that influence the fate and function of MSCs. Additionally, the new discoveries and suggestions regarding their molecular mechanisms will be explained.
Collapse
Affiliation(s)
| | - Elham Alimoradi
- Department of biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Hassan Akrami
- Department of biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
262
|
Crowder SW, Balikov DA, Boire TC, McCormack D, Lee JB, Gupta MK, Skala MC, Sung HJ. Copolymer-Mediated Cell Aggregation Promotes a Proangiogenic Stem Cell Phenotype In Vitro and In Vivo. Adv Healthc Mater 2016; 5:2866-2871. [PMID: 27717208 PMCID: PMC5152909 DOI: 10.1002/adhm.201600819] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/19/2016] [Indexed: 12/31/2022]
Abstract
Material-induced cell aggregation drives a proangiogenic expression profile. Copolymer substrates containing cell-repellent and cell-adhesive domains force the aggregation of human mesenchymal stem cells, which results in enhanced tubulogenesis in vitro and stabilization of vasculature in vivo. These findings can be used to design instructive biomaterial scaffolds for clinical use.
Collapse
Affiliation(s)
- Spencer W. Crowder
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Daniel A. Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Timothy C. Boire
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Devin McCormack
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jung Bok Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
263
|
Liu Y, Muñoz N, Tsai AC, Logan TM, Ma T. Metabolic Reconfiguration Supports Reacquisition of Primitive Phenotype in Human Mesenchymal Stem Cell Aggregates. Stem Cells 2016; 35:398-410. [DOI: 10.1002/stem.2510] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/16/2016] [Accepted: 08/21/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Yijun Liu
- Department of Chemical and Biomedical Engineering; Florida State University; Tallahassee Florida USA
| | - Nathalie Muñoz
- Graduate Program in Molecular Biophysics, Florida State University; Tallahassee Florida USA
| | - Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering; Florida State University; Tallahassee Florida USA
| | - Timothy M. Logan
- Graduate Program in Molecular Biophysics, Florida State University; Tallahassee Florida USA
- Department of Chemistry and Biochemistry; Florida State University; Tallahassee Florida USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering; Florida State University; Tallahassee Florida USA
- Graduate Program in Molecular Biophysics, Florida State University; Tallahassee Florida USA
| |
Collapse
|
264
|
Bartosh TJ, Ullah M, Zeitouni S, Beaver J, Prockop DJ. Cancer cells enter dormancy after cannibalizing mesenchymal stem/stromal cells (MSCs). Proc Natl Acad Sci U S A 2016; 113:E6447-E6456. [PMID: 27698134 PMCID: PMC5081643 DOI: 10.1073/pnas.1612290113] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Patients with breast cancer often develop malignant regrowth of residual drug-resistant dormant tumor cells years after primary treatment, a process defined as cancer relapse. Deciphering the causal basis of tumor dormancy therefore has obvious therapeutic significance. Because cancer cell behavior is strongly influenced by stromal cells, particularly the mesenchymal stem/stromal cells (MSCs) that are actively recruited into tumor-associated stroma, we assessed the impact of MSCs on breast cancer cell (BCC) dormancy. Using 3D cocultures to mimic the cellular interactions of an emerging tumor niche, we observed that MSCs sequentially surrounded the BCCs, promoted formation of cancer spheroids, and then were internalized/degraded through a process resembling the well-documented yet ill-defined clinical phenomenon of cancer cell cannibalism. This suspected feeding behavior was less appreciable in the presence of a rho kinase inhibitor and in 2D monolayer cocultures. Notably, cannibalism of MSCs enhanced survival of BCCs deprived of nutrients but suppressed their tumorigenicity, together suggesting the cancer cells entered dormancy. Transcriptome profiles revealed that the resulting BCCs acquired a unique molecular signature enriched in prosurvival factors and tumor suppressors, as well as inflammatory mediators that demarcate the secretome of senescent cells, also referred to as the senescence-associated secretory phenotype. Overall, our results provide intriguing evidence that cancer cells under duress enter dormancy after cannibalizing MSCs. Importantly, our practical 3D coculture model could provide a valuable tool to understand the antitumor activity of MSCs and cell cannibalism further, and therefore open new therapeutic avenues for the prevention of cancer recurrence.
Collapse
Affiliation(s)
- Thomas J Bartosh
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76502; Medical Physiology, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76504
| | - Mujib Ullah
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76502
| | - Suzanne Zeitouni
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76502
| | - Joshua Beaver
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76502; Medical Physiology, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76504
| | - Darwin J Prockop
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University System Health Science Center, Temple, TX 76502;
| |
Collapse
|
265
|
Bang OY, Kim EH, Cha JM, Moon GJ. Adult Stem Cell Therapy for Stroke: Challenges and Progress. J Stroke 2016; 18:256-266. [PMID: 27733032 PMCID: PMC5066440 DOI: 10.5853/jos.2016.01263] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/15/2016] [Accepted: 09/18/2016] [Indexed: 02/06/2023] Open
Abstract
Stroke is one of the leading causes of death and physical disability among adults. It has been 15 years since clinical trials of stem cell therapy in patients with stroke have been conducted using adult stem cells like mesenchymal stem cells and bone marrow mononuclear cells. Results of randomized controlled trials showed that adult stem cell therapy was safe but its efficacy was modest, underscoring the need for new stem cell therapy strategies. The primary limitations of current stem cell therapies include (a) the limited source of engraftable stem cells, (b) the presence of optimal time window for stem cell therapies, (c) inherited limitation of stem cells in terms of growth, trophic support, and differentiation potential, and (d) possible transplanted cell-mediated adverse effects, such as tumor formation. Here, we discuss recent advances that overcome these hurdles in adult stem cell therapy for stroke.
Collapse
Affiliation(s)
- Oh Young Bang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Eun Hee Kim
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Jae Min Cha
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Seoul, Korea.,Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Gyeong Joon Moon
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea.,Stem cell and Regenerative Medicine Institute, Samsung Biomedical Research Institute, Seoul, Korea
| |
Collapse
|
266
|
Chung TW, Lo HY, Chou TH, Chen JH, Wang SS. Promoting Cardiomyogenesis of hBMSC with a Forming Self-Assembly hBMSC Microtissues/HA-GRGD/SF-PCL Cardiac Patch Is Mediated by the Synergistic Functions of HA-GRGD. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/18/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Tze-Wen Chung
- Department of Biomedical Engineering; National Yang-Ming University; Taipei 11221 Taiwan
| | - Hsin-Yu Lo
- Department of Biomedical Engineering; National Yang-Ming University; Taipei 11221 Taiwan
| | - Tzung-Han Chou
- Department of Chemical Engineering; National Yunlin University of Science and Technology; Yunlin 64402 Taiwan
| | - Jan-Hou Chen
- Department of Chemical Engineering; National Yunlin University of Science and Technology; Yunlin 64402 Taiwan
| | - Shoei-Shen Wang
- Department of SurgeryNational Taiwan University Hospital; National Taiwan University College of Medicine; Taipei 110 Taiwan
| |
Collapse
|
267
|
Regmi S, Jeong JH. Superiority of three-dimensional stem cell clusters over monolayer culture: An archetype to biological application. Macromol Res 2016. [DOI: 10.1007/s13233-016-4107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
268
|
de Soure AM, Fernandes-Platzgummer A, da Silva CL, Cabral JMS. Scalable microcarrier-based manufacturing of mesenchymal stem/stromal cells. J Biotechnol 2016; 236:88-109. [PMID: 27527397 DOI: 10.1016/j.jbiotec.2016.08.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/02/2016] [Accepted: 08/09/2016] [Indexed: 12/17/2022]
Abstract
Due to their unique features, mesenchymal stem/stromal cells (MSC) have been exploited in clinical settings as therapeutic candidates for the treatment of a variety of diseases. However, the success in obtaining clinically-relevant MSC numbers for cell-based therapies is dependent on efficient isolation and ex vivo expansion protocols, able to comply with good manufacturing practices (GMP). In this context, the 2-dimensional static culture systems typically used for the expansion of these cells present several limitations that may lead to reduced cell numbers and compromise cell functions. Furthermore, many studies in the literature report the expansion of MSC using fetal bovine serum (FBS)-supplemented medium, which has been critically rated by regulatory agencies. Alternative platforms for the scalable manufacturing of MSC have been developed, namely using microcarriers in bioreactors, with also a considerable number of studies now reporting the production of MSC using xenogeneic/serum-free medium formulations. In this review we provide a comprehensive overview on the scalable manufacturing of human mesenchymal stem/stromal cells, depicting the various steps involved in the process from cell isolation to ex vivo expansion, using different cell tissue sources and culture medium formulations and exploiting bioprocess engineering tools namely microcarrier technology and bioreactors.
Collapse
Affiliation(s)
- António M de Soure
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa, Portugal.
| |
Collapse
|
269
|
Lei J, Trevino E, Temenoff J. Cell number and chondrogenesis in human mesenchymal stem cell aggregates is affected by the sulfation level of heparin used as a cell coating. J Biomed Mater Res A 2016; 104:1817-29. [PMID: 26990913 PMCID: PMC5532474 DOI: 10.1002/jbm.a.35713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 01/12/2023]
Abstract
For particular cell-based therapies, it may be required to culture mesenchymal stem cell (MSC) aggregates with growth factors to promote cell proliferation and/or differentiation. Heparin, a negatively charged glycosaminoglycan (GAG) is known to play an important role in sequestration of positively charged growth factors and, when incorporated within cellular aggregates, could be used to promote local availability of growth factors. We have developed a heparin-based cell coating and we believe that the electrostatic interaction between native heparin and the positively charged growth factors will result in (1) higher cell number in response to fibroblast growth factor-2 (FGF-2) and 2) greater chondrogenic differentiation in response to transforming growth factor-β1 (TGF-β1), compared to a desulfated heparin coating. Results revealed that in the presence of FGF-2, by day 14, heparin-coated MSC aggregates increased in DNA content 8.5 ± 1.6 fold compared to day 1, which was greater than noncoated and desulfated heparin-coated aggregates. In contrast, when cultured in the presence of TGF-β1, by day 21, desulfated heparin-coated aggregates upregulated gene expression of collagen II by 86.5 ± 7.5 fold and collagen X by 37.1 ± 4.7 fold, which was higher than that recorded in the noncoated and heparin-coated aggregates. These observations indicate that this coating technology represents a versatile platform to design MSC culture systems with pairings of GAGs and growth factors that can be tailored to overcome specific challenges in scale-up and culture for MSC-based therapeutics. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1817-1829, 2016.
Collapse
Affiliation(s)
- Jennifer Lei
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, 30332, Georgia
| | - Elda Trevino
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, Georgia
| | - Johnna Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, Georgia
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, 30332, Georgia
| |
Collapse
|
270
|
Murphy KC, Hoch AI, Harvestine JN, Zhou D, Leach JK. Mesenchymal Stem Cell Spheroids Retain Osteogenic Phenotype Through α2β1 Signaling. Stem Cells Transl Med 2016; 5:1229-37. [PMID: 27365484 PMCID: PMC4996446 DOI: 10.5966/sctm.2015-0412] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/07/2016] [Indexed: 12/22/2022] Open
Abstract
The induction of mesenchymal stem cells (MSCs) toward the osteoblastic lineage using osteogenic supplements prior to implantation is one approach under examination to enhance their bone-forming potential. Spheroids formed from induced cells exhibited improved retention of osteogenic markers as a function of integrin binding to cell-secreted extracellular matrix (ECM). These results demonstrate the capacity of spheroidal culture to sustain the mineral-producing phenotype of MSCs, thus enhancing their contribution toward bone formation and repair. The induction of mesenchymal stem cells (MSCs) toward the osteoblastic lineage using osteogenic supplements prior to implantation is one approach under examination to enhance their bone-forming potential. MSCs rapidly lose their induced phenotype upon removal of the soluble stimuli; however, their bone-forming potential can be sustained when provided with continued instruction via extracellular matrix (ECM) cues. In comparison with dissociated cells, MSC spheroids exhibit improved survival and secretion of trophic factors while maintaining their osteogenic potential. We hypothesized that entrapment of MSC spheroids formed from osteogenically induced cells would exhibit better preservation of their bone-forming potential than would dissociated cells from monolayer culture. Spheroids exhibited comparable osteogenic potential and increased proangiogenic potential with or without osteogenic preconditioning versus monolayer-cultured MSCs. Spheroids were then entrapped in collagen hydrogels, and the osteogenic stimulus was removed. In comparison with entrapped dissociated MSCs, spheroids exhibited significantly increased markers of osteogenic differentiation. The capacity of MSC spheroids to retain their osteogenic phenotype upon withdrawal of inductive cues was mediated by α2β1 integrin binding to cell-secreted ECM. These results demonstrate the capacity of spheroidal culture to sustain the mineral-producing phenotype of MSCs, thus enhancing their contribution toward bone formation and repair. Significance Despite the promise of mesenchymal stem cells (MSCs) for cell-based therapies for tissue repair and regeneration, there is little evidence that transplanted MSCs directly contribute to new bone formation, suggesting that induced cells rapidly lose their osteogenic phenotype or undergo apoptosis. In comparison with dissociated cells, MSC spheroids exhibit increased trophic factor secretion and improved cell survival. The loss of phenotype represents a significant clinical challenge for cell therapies, yet there is no evidence for whether MSC spheroids retain their osteogenic phenotype upon entrapment in a clinically relevant biomaterial. These findings demonstrate that MSC spheroids retain their osteogenic phenotype better than do dissociated MSCs, and this is due to integrin engagement with the cell-secreted extracellular matrix. These data provide evidence for a novel approach for potentiating the use of MSCs in bone repair.
Collapse
Affiliation(s)
- Kaitlin C Murphy
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Allison I Hoch
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Jenna N Harvestine
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Dejie Zhou
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
271
|
Obara C, Tomiyama KI, Takizawa K, Islam R, Yasuda T, Gotoh T, Tajima K. Characteristics of three-dimensional prospectively isolated mouse bone marrow mesenchymal stem/stromal cell aggregates on nanoculture plates. Cell Tissue Res 2016; 366:113-27. [DOI: 10.1007/s00441-016-2405-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 04/04/2016] [Indexed: 01/01/2023]
|
272
|
Bellotti C, Duchi S, Bevilacqua A, Lucarelli E, Piccinini F. Long term morphological characterization of mesenchymal stromal cells 3D spheroids built with a rapid method based on entry-level equipment. Cytotechnology 2016; 68:2479-2490. [PMID: 27023795 DOI: 10.1007/s10616-016-9969-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/22/2016] [Indexed: 11/25/2022] Open
Abstract
Three-dimensional (3D) spheroids of mesenchymal stromal cells (MSC) have been demonstrated to improve a wide range of MSC features, such as multilineage potential, secretion of therapeutic factors, and resistance against hypoxic condition. Accordingly, they represent a promising tool in regenerative medicine for several biological and clinical applications. Many approaches have been proposed to generate MSC spheroids. They usually require specific generation systems, such as rotatory bioreactors or low-attachment plates, and each approach has its own disadvantages. Furthermore, an over-time analysis of morphological homogeneity and architectural stability of the spheroids generated is rarely provided. In this work we adapted the "pellet culture" method to obtain homogenous spheroids of MSC and maintain them in vitro for long term studies. We analysed their outer and inner structure over a 2-month period to provide morphological and architectural information regarding the spheroids generated. Quantitative and qualitative data were obtained using brightfield and confocal microscope imaging coupled to a computational analysis to estimate volume, sphericity, and jagging degree. In addition, histological evaluation was performed to more thoroughly assess the cellular composition and the internal architecture of the 3D spheroids. The results provided show that MSC spheroids generated with the proposed approach are homogeneous and stable, from both morphological and architectural points of view, for a period of at least 15 days, approximately between day 15 and day 30 after their generation. Accordingly, the approach proposed serves as a rapid, cost-effective, and efficient method to generate and maintain MSC spheroids using common entry-level laboratory equipment only.
Collapse
Affiliation(s)
- Chiara Bellotti
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Serena Duchi
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Alessandro Bevilacqua
- Advanced Research Center on Electronic Systems for Information and Communication Technologies "E. De Castro" (ARCES), University of Bologna, Via Toffano 2/2, 40125, Bologna, Italy.,Department of Computer Science and Engineering (DISI), University of Bologna, Bologna, Italy
| | - Enrico Lucarelli
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Filippo Piccinini
- Advanced Research Center on Electronic Systems for Information and Communication Technologies "E. De Castro" (ARCES), University of Bologna, Via Toffano 2/2, 40125, Bologna, Italy.
| |
Collapse
|
273
|
Self-patterning of adipose-derived mesenchymal stem cells and chondrocytes cocultured on hyaluronan-grafted chitosan surface. Biointerphases 2016; 11:011011. [PMID: 26916660 DOI: 10.1116/1.4942754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The articular cartilage, once injured, has a limited capacity for intrinsic repair. Preparation of functionally biocartilage substitutes in vitro for cartilage repair is an attractive concept with the recent advances in tissue engineering. In this study, adipose-derived adult stem cells (ADAS) and chondrocytes (Ch) were cocultured in different population ratios on the surface of hyaluronan-grafted chitosan (CS-HA) membranes. The two types of cells could self-assemble into cospheroids with different morphologies. In particular, when ADAS and Ch were cocultured at an initial ratio of 7:3 on CS-HA surface, the expression of chondrogenic markers was upregulated, leading to preferred chondrogenesis of the cospheroids. Therefore, using the ADAS/Ch 7:3 cospheroids derived on CS-HA surface instead of using only a single type of cells may be favorable for future therapeutic applications.
Collapse
|
274
|
Cesarz Z, Funnell JL, Guan J, Tamama K. Soft Elasticity-Associated Signaling and Bone Morphogenic Protein 2 Are Key Regulators of Mesenchymal Stem Cell Spheroidal Aggregates. Stem Cells Dev 2016; 25:622-35. [PMID: 26916040 DOI: 10.1089/scd.2015.0356] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell therapy with adult mesenchymal stem cells (MSCs) is a promising approach to regenerative medicine and autoimmune diseases. There are various approaches to improve the efficacy of MSC-based therapeutics, and MSC preparation as spheroidal aggregates, or MSC spheroids, is a novel preparatory and delivery method. Spheroid formation induces a dramatic change in the gene expression profile of MSCs. Self-activation of interleukin-1 (IL1) signaling was shown to be upstream of both pro- and anti-inflammatory genes in MSC spheroids, but the molecular pathways that initiate IL1 signaling remain unknown. As bone morphogenic protein (BMP)2 upregulation precedes that of IL1B expression during spheroid formation, we hypothesized that BMP2 signaling triggers IL1 signaling in MSC spheroids. Contrary to expectations, BMP2 signaling decreased expression of IL1B and downstream genes in a SMAD6-dependent manner. Conversely, IL1B signaling enhanced BMP2 expression. Another major difference between two-dimensional (2D) monolayer culture and three-dimensional (3D) spheroid culture is the Young's elasticity modulus, or stiffness, of the materials surrounding the cells, as there is a million-fold difference between a plastic surface for standard 2D culture (GPa) and 3D spheroidal aggregates (0.1 kPa). We tested another hypothesis that soft elasticity-associated mechano-signaling initiates the gene expression change during spheroid formation. Results showed that both BMP2 expression and inflammatory signaling are upregulated in an elasticity-associated signaling-dependent manner in MSCs. Lastly, BMP2 signaling enhanced cell survival and cell spreading of MSC spheroids. In summary, our study suggests that soft elasticity and BMP2 signaling are critical for MSC spheroids.
Collapse
Affiliation(s)
- Zoe Cesarz
- 1 Department of Pathology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Jessica L Funnell
- 1 Department of Pathology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Jianjun Guan
- 2 Department of Materials Science and Engineering, the Ohio State University , Columbus, Ohio
| | - Kenichi Tamama
- 1 Department of Pathology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
275
|
Microtissues in Cardiovascular Medicine: Regenerative Potential Based on a 3D Microenvironment. Stem Cells Int 2016; 2016:9098523. [PMID: 27073399 PMCID: PMC4814701 DOI: 10.1155/2016/9098523] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023] Open
Abstract
More people die annually from cardiovascular diseases than from any other cause. In particular, patients who suffer from myocardial infarction may be affected by ongoing adverse remodeling processes of the heart that may ultimately lead to heart failure. The introduction of stem and progenitor cell-based applications has raised substantial hope for reversing these processes and inducing cardiac regeneration. However, current stem cell therapies using single-cell suspensions have failed to demonstrate long-lasting efficacy due to the overall low retention rate after cell delivery to the myocardium. To overcome this obstacle, the concept of 3D cell culture techniques has been proposed to enhance therapeutic efficacy and cell engraftment based on the simulation of an in vivo-like microenvironment. Of great interest is the use of so-called microtissues or spheroids, which have evolved from their traditional role as in vitro models to their novel role as therapeutic agents. This review will provide an overview of the therapeutic potential of microtissues by addressing primarily cardiovascular regeneration. It will accentuate their advantages compared to other regenerative approaches and summarize the methods for generating clinically applicable microtissues. In addition, this review will illustrate the unique properties of the microenvironment within microtissues that makes them a promising next-generation therapeutic approach.
Collapse
|
276
|
Follin B, Juhl M, Cohen S, Pedersen AE, Kastrup J, Ekblond A. Increased Paracrine Immunomodulatory Potential of Mesenchymal Stromal Cells in Three-Dimensional Culture. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:322-9. [PMID: 26861485 PMCID: PMC4964752 DOI: 10.1089/ten.teb.2015.0532] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have been investigated extensively through the past years, proving to have great clinical therapeutic potential. In vitro cultivation of MSCs in three-dimensional (3D) culture systems, such as scaffolds, hydrogels, or spheroids, have recently gained attention for tissue engineering applications. Studies on MSC spheroids demonstrated that such cultivation increased the paracrine immunomodulatory potential of the MSCs, accompanied by phenotypic alterations. In this review, we gather results from recent experimental studies on the immunomodulatory abilities of MSCs when cultured as spheroids or in biomaterials like scaffolds or hydrogels compared to regular two-dimensional (2D) culture and show that alterations occurring to MSCs in spheroids also occur in MSCs in biomaterials. We provide a brief description of known mechanisms of MSC immunomodulatory capacity and how they are altered in the two 3D culture systems, together with phenotypic cellular changes. Based on the present knowledge, we highlight vital areas in need of further investigation. The impact of 3D environments on immunomodulation has great potential for tissue engineering and cellular therapy, and this is the first review to gather this knowledge with a comparison across different 3D environments.
Collapse
Affiliation(s)
- Bjarke Follin
- 1 Cardiology Stem Cell Center, The Heart Center, Rigshospitalet, Copenhagen University Hospital , Copenhagen, Denmark
| | - Morten Juhl
- 1 Cardiology Stem Cell Center, The Heart Center, Rigshospitalet, Copenhagen University Hospital , Copenhagen, Denmark
| | - Smadar Cohen
- 2 Regenerative Medicine and Stem Cell Research Center, the Avram and Stella Goldstein-Goren Department of Biotechnology and Engineering, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Anders Elm Pedersen
- 3 Department of Immunology and Microbiology, University of Copenhagen , Copenhagen, Denmark
| | - Jens Kastrup
- 1 Cardiology Stem Cell Center, The Heart Center, Rigshospitalet, Copenhagen University Hospital , Copenhagen, Denmark
| | - Annette Ekblond
- 1 Cardiology Stem Cell Center, The Heart Center, Rigshospitalet, Copenhagen University Hospital , Copenhagen, Denmark
| |
Collapse
|
277
|
Lu S, Lee EJ, Lam J, Tabata Y, Mikos AG. Evaluation of Gelatin Microparticles as Adherent-Substrates for Mesenchymal Stem Cells in a Hydrogel Composite. Ann Biomed Eng 2016; 44:1894-907. [PMID: 26935924 DOI: 10.1007/s10439-016-1582-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 02/26/2016] [Indexed: 12/14/2022]
Abstract
Due to the lack of cell-adhesive moieties in traditional synthetic hydrogels, the present work investigated the use of degradable gelatin microparticles (GMPs) as temporary adherent substrates for anchorage-dependent mesenchymal stem cells (MSCs). MSCs were seeded onto GMPs of varying crosslinking densities and sizes to investigate their role on influencing MSC differentiation and aggregation. The MSC-seeded GMPs were then encapsulated in poly(ethylene glycol)-based hydrogels and cultured in serum-free, growth factor-free osteochondral medium. Non-seeded MSCs co-encapsulated with GMPs in the hydrogels were used as a control for comparison. Over the course of 35 days, MSCs seeded on GMPs exhibited more cell-cell contacts, greater chondrogenic potential, and a down-regulation of osteogenic markers compared to the controls. Although the factors of GMP crosslinking and size had nominal influence on MSC differentiation and aggregation, GMPs demonstrate potential as an adherent-substrate for improving cell delivery from hydrogel scaffolds by facilitating cell-cell contacts and improving MSC differentiation.
Collapse
Affiliation(s)
- Steven Lu
- Department of Bioengineering, Rice University, P.O. Box 1892, MS-142, Houston, TX, 77005-1892, USA
| | - Esther J Lee
- Department of Bioengineering, Rice University, P.O. Box 1892, MS-142, Houston, TX, 77005-1892, USA
| | - Johnny Lam
- Department of Bioengineering, Rice University, P.O. Box 1892, MS-142, Houston, TX, 77005-1892, USA
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, P.O. Box 1892, MS-142, Houston, TX, 77005-1892, USA.
| |
Collapse
|
278
|
Ozdemir T, Fowler EW, Hao Y, Ravikrishnan A, Harrington DA, Witt RL, Farach-Carson MC, Pradhan-Bhatt S, Jia X. Biomaterials-based strategies for salivary gland tissue regeneration. Biomater Sci 2016; 4:592-604. [PMID: 26878077 DOI: 10.1039/c5bm00358j] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The salivary gland is a complex, secretory tissue that produces saliva and maintains oral homeostasis. Radiation induced salivary gland atrophy, manifested as "dry mouth" or xerostomia, poses a significant clinical challenge. Tissue engineering recently has emerged as an alternative, long-term treatment strategy for xerostomia. In this review, we summarize recent efforts towards the development of functional and implantable salivary glands utilizing designed polymeric substrates or synthetic matrices/scaffolds. Although the in vitro engineering of a complex implantable salivary gland is technically challenging, opportunities exist for multidisciplinary teams to assemble implantable and secretory tissue modules by combining stem/progenitor cells found in the adult glands with biomimetic and cell-instructive materials.
Collapse
Affiliation(s)
- Tugba Ozdemir
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Hong S, Lee JY, Hwang C, Shin JH, Park Y. Inhibition of Rho-Associated Protein Kinase Increases the Angiogenic Potential of Mesenchymal Stem Cell Aggregates via Paracrine Effects. Tissue Eng Part A 2016; 22:233-43. [PMID: 26592750 DOI: 10.1089/ten.tea.2015.0289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The aggregation of multiple cells, such as mesenchymal condensation, is an important biological process in skeletal muscle development, osteogenesis, and adipogenesis. Due to limited in vivo study model systems, a simple and effective in vitro three-dimensional (3D) aggregation system is required to study the mechanisms of multicellular aggregation and its applications. We first generated controlled mesenchymal stem cell (MSC) aggregates using a bioprinting technique to monitor their aggregation and sprouting. We induced the angiogenic potential of the MSCs through chemical inhibition of the Rho/Rho-associated protein kinase (ROCK) pathway, which led to hairy sprouting in the aggregates. The angiogenic potential of this 3D construct was then tested by subcutaneously implanting the Matrigel with 3D MSC aggregates in a rat. Treatment of 3D MSCs with the ROCK inhibitor, Y27632, increased their angiogenic activity in vivo. The gene expressions and histological staining indicated that angiogenesis and neovascularization were mainly regulated by the paracrine factors secreted from human 3D MSC constructs. Our results demonstrate the enhancement of the angiogenic potential of the MSC constructs through the secretion of vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) by the inhibition of the Rho/ROCK pathway.
Collapse
Affiliation(s)
- Soyoung Hong
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea.,2 Biomedical Engineering Research Center, Asan Medical Center , Seoul, Korea
| | - Jae Yeon Lee
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea
| | - Changmo Hwang
- 2 Biomedical Engineering Research Center, Asan Medical Center , Seoul, Korea
| | - Jennifer H Shin
- 3 Department of Mechanical Engineering, Graduate School of Medical Science and Engineering , KAIST, Daejeon, Korea
| | - Yongdoo Park
- 1 Department of Biomedical Engineering, College of Medicine, Korea University , Seoul, Korea
| |
Collapse
|
280
|
Sridharan B, Lin SM, Hwu AT, Laflin AD, Detamore MS. Stem Cells in Aggregate Form to Enhance Chondrogenesis in Hydrogels. PLoS One 2015; 10:e0141479. [PMID: 26719986 PMCID: PMC4697858 DOI: 10.1371/journal.pone.0141479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
There are a variety of exciting hydrogel technologies being explored for cartilage regenerative medicine. Our overall goal is to explore whether using stem cells in an aggregate form may be advantageous in these applications. 3D stem cell aggregates hold great promise as they may recapitulate the in vivo skeletal tissue condensation, a property that is not typically observed in 2D culture. We considered two different stem cell sources, human umbilical cord Wharton’s jelly cells (hWJCs, currently being used in clinical trials) and rat bone marrow-derived mesenchymal stem cells (rBMSCs). The objective of the current study was to compare the influence of cell phenotype, aggregate size, and aggregate number on chondrogenic differentiation in a generic hydrogel (agarose) platform. Despite being differing cell sources, both rBMSC and hWJC aggregates were consistent in outperforming cell suspension control groups in biosynthesis and chondrogenesis. Higher cell density impacted biosynthesis favorably, and the number of aggregates positively influenced chondrogenesis. Therefore, we recommend that investigators employing hydrogels consider using cells in an aggregate form for enhanced chondrogenic performance.
Collapse
Affiliation(s)
- BanuPriya Sridharan
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
| | - Staphany M. Lin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Alexander T. Hwu
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Amy D. Laflin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Michael S. Detamore
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
281
|
Sart S, Agathos SN, Li Y, Ma T. Regulation of mesenchymal stem cell 3D microenvironment: From macro to microfluidic bioreactors. Biotechnol J 2015; 11:43-57. [PMID: 26696441 DOI: 10.1002/biot.201500191] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 11/02/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have emerged as an important cell type in cell therapy and tissue engineering. In these applications, maintaining the therapeutic properties of hMSCs requires tight control of the culture environments and the structural cell organizations. Bioreactor systems are essential tools to achieve these goals in the clinical-scale expansion and tissue engineering applications. This review summarizes how different bioreactors provide cues to regulate the structure and the chemico-mechanical microenvironment of hMSCs with a focus on 3D organization. In addition to conventional bioreactors, recent advances in microfluidic bioreactors as a novel approach to better control the hMSC microenvironment are also discussed. These advancements highlight the key role of bioreactor systems in preserving hMSC's functional properties by providing dynamic and temporal regulation of in vitro cellular microenvironment.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory, CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Spiros N Agathos
- Laboratory of Bioengineering, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA.
| |
Collapse
|
282
|
Hutton DL, Grayson WL. Hypoxia Inhibits De Novo Vascular Assembly of Adipose-Derived Stromal/Stem Cell Populations, but Promotes Growth of Preformed Vessels. Tissue Eng Part A 2015; 22:161-9. [PMID: 26481655 DOI: 10.1089/ten.tea.2015.0421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Vascularization is critical for cell survival within tissue-engineered grafts. Adipose-derived stromal/stem cells (ASCs) are widely used in tissue engineering applications as they are a clinically relevant source of stem cells and endothelial progenitor cells. ASCs have previously been shown to self-assemble into pericyte-stabilized vascular networks in normoxic (20% O2) cultures. This capacity for de novo vascular assembly may accelerate graft vascularization in vivo rather than relying solely on angiogenic ingrowth. However, oxygen depletion within large cell-seeded grafts will be rapid, and it is unclear how this worsening hypoxic environment will impact the vascular assembly of the transplanted cells. The objectives of this study were to determine whether ASC-derived vessels could grow in hypoxia and to assess whether the vessel maturity (i.e., individual cells vs. preformed vessels) influenced this hypoxic response. Utilizing an in vitro vascularization model, ASCs were encapsulated within fibrin gels and cultured in vitro for up to 6 days in either normoxia (20% O2) or hypoxia (0.2% or 2% O2). In a subsequent experiment, vessels were allowed to preform in normoxia for 6 days before an additional 6 days of either normoxia or hypoxia. Viability, vessel growth, pericyte coverage, proliferation, metabolism, and angiogenic factor expression were assessed for each experimental approach. Vessel growth was dramatically inhibited in both moderate and severe hypoxia (47% and 11% total vessel length vs. normoxia, respectively), despite maintaining high cell viability and upregulating endogenous expression of vascular endothelial growth factor in hypoxia. Bromodeoxyuridine labeling indicated significantly reduced proliferation of endothelial cells in hypoxia. In contrast, when vascular networks were allowed to preform for 6 days in normoxia, vessels not only survived but also continued to grow more in hypoxia than those maintained in normoxia. These findings demonstrate that vascular assembly and growth are tightly regulated by oxygen tension and may be differentially affected by hypoxic conditions based on the maturity of the vessels. Understanding this relationship is critical to developing effective approaches to engineer viable tissue-engineered grafts in vivo.
Collapse
Affiliation(s)
- Daphne L Hutton
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Warren L Grayson
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Department of Material Sciences and Engineering, Johns Hopkins University School of Engineering , Baltimore, Maryland
| |
Collapse
|
283
|
Panchalingam KM, Jung S, Rosenberg L, Behie LA. Bioprocessing strategies for the large-scale production of human mesenchymal stem cells: a review. Stem Cell Res Ther 2015; 6:225. [PMID: 26597928 PMCID: PMC4657237 DOI: 10.1186/s13287-015-0228-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs), also called mesenchymal stromal cells, have been of great interest in regenerative medicine applications because of not only their differentiation potential but also their ability to secrete bioactive factors that can modulate the immune system and promote tissue repair. This potential has initiated many early-phase clinical studies for the treatment of various diseases, disorders, and injuries by using either hMSCs themselves or their secreted products. Currently, hMSCs for clinical use are generated through conventional static adherent cultures in the presence of fetal bovine serum or human-sourced supplements. However, these methods suffer from variable culture conditions (i.e., ill-defined medium components and heterogeneous culture environment) and thus are not ideal procedures to meet the expected future demand of quality-assured hMSCs for human therapeutic use. Optimizing a bioprocess to generate hMSCs or their secreted products (or both) promises to improve the efficacy as well as safety of this stem cell therapy. In this review, current media and methods for hMSC culture are outlined and bioprocess development strategies discussed.
Collapse
Affiliation(s)
- Krishna M Panchalingam
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Sunghoon Jung
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Lawrence Rosenberg
- Department of Surgery, McGill University Health Centre, 845 Rue Sherbrooke Quest, Montreal, QC, H3G 1A4, Canada.,Jewish General Hospital, 3755 Chemin de la Côte-Ste-Catherine Road, Montreal, QC, H3T 1E2, Canada
| | - Leo A Behie
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
284
|
Abstract
Compared with traditional 2D adherent cell culture, 3D spheroidal cell aggregates, or spheroids, are regarded as more physiological, and this technique has been exploited in the field of oncology, stem cell biology, and tissue engineering. Mesenchymal stem cells (MSCs) cultured in spheroids have enhanced anti-inflammatory, angiogenic, and tissue reparative/regenerative effects with improved cell survival after transplantation. Cytoskeletal reorganization and drastic changes in cell morphology in MSC spheroids indicate a major difference in mechanophysical properties compared with 2D culture. Enhanced multidifferentiation potential, upregulated expression of pluripotency marker genes, and delayed replicative senescence indicate enhanced stemness in MSC spheroids. Furthermore, spheroid formation causes drastic changes in the gene expression profile of MSC in microarray analyses. In spite of these significant changes, underlying molecular mechanisms and signaling pathways triggering and sustaining these changes are largely unknown.
Collapse
|
285
|
Three-Dimensional Aggregates Enhance the Therapeutic Effects of Adipose Mesenchymal Stem Cells for Ischemia-Reperfusion Induced Kidney Injury in Rats. Stem Cells Int 2015; 2016:9062638. [PMID: 26649053 PMCID: PMC4663369 DOI: 10.1155/2016/9062638] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/27/2015] [Indexed: 01/09/2023] Open
Abstract
It has been shown that administration of adipose derived mesenchymal stem cells (AdMSCs) enhanced structural and functional recovery of renal ischemia-reperfusion (IR) injury. Low engraftment of stem cells, however, limits the therapeutic effects of AdMSCs. The present study was designed to enhance the therapeutic effects of AdMSCs by delivering AdMSCs in a three-dimensional (3D) aggregates form. Microwell was used to produce 3D AdMSCs aggregates. In vitro data indicated that AdMSCs in 3D aggregates were less susceptible to oxidative and hypoxia stress induced by 200 μM peroxide and hypoxia/reoxygenation, respectively, compared with those cultured in two-dimensional (2D) monolayer. Furthermore, AdMSCs in 3D aggregates secreted more proangiogenic factors than those cultured in 2D monolayer. 2D AdMSCs or 3D AdMSCs aggregates were injected into renal cortex immediately after induction of renal IR injury. In vivo data revealed that 3D aggregates enhanced the effects of AdMSCs in recovering function and structure after renal IR injury. Improved grafted AdMSCs were observed in kidney injected with 3D aggregates compared with AdMSCs cultured in 2D monolayer. Our results demonstrated that 3D AdMSCs aggregated produced by microwell enhanced the retention and therapeutic effects of AdMSCs for renal IR injury.
Collapse
|
286
|
Hofmeister LH, Costa L, Balikov DA, Crowder SW, Terekhov A, Sung HJ, Hofmeister WH. Patterned polymer matrix promotes stemness and cell-cell interaction of adult stem cells. J Biol Eng 2015; 9:18. [PMID: 26464581 PMCID: PMC4603908 DOI: 10.1186/s13036-015-0016-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/21/2015] [Indexed: 12/27/2022] Open
Abstract
Background The interaction of stem cells with their culture substrates is critical in controlling their fate and function. Declining stemness of adult-derived human mesenchymal stem cells (hMSCs) during in vitro expansion on tissue culture polystyrene (TCPS) severely limits their therapeutic efficacy prior to cell transplantation into damaged tissues. Thus, various formats of natural and synthetic materials have been manipulated in attempts to reproduce in vivo matrix environments in which hMSCs reside. Results We developed a series of patterned polymer matrices for cell culture by hot-pressing poly(ε-caprolactone) (PCL) films in femtosecond laser-ablated nanopore molds, forming nanofibers on flat PCL substrates. hMSCs cultured on these PCL fiber matrices significantly increased expression of critical self-renewal factors, Nanog and OCT4A, as well as markers of cell-cell interaction PECAM and ITGA2. The results suggest the patterned polymer fiber matrix is a promising model to maintain the stemness of adult hMSCs. Conclusion This approach meets the need for scalable, highly repeatable, and tuneable models that mimic extracellular matrix features that signal for maintenance of hMSC stemness.
Collapse
Affiliation(s)
- Lucas H Hofmeister
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - Lino Costa
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN USA
| | - Daniel A Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - Spencer W Crowder
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - Alexander Terekhov
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN USA
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
| | - William H Hofmeister
- Center for Laser Applications, University of Tennessee Space Institute, Tullahoma, TN USA
| |
Collapse
|
287
|
Controlling Redox Status for Stem Cell Survival, Expansion, and Differentiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:105135. [PMID: 26273419 PMCID: PMC4530287 DOI: 10.1155/2015/105135] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/06/2014] [Indexed: 01/07/2023]
Abstract
Reactive oxygen species (ROS) have long been considered as pathological agents inducing apoptosis under adverse culture conditions. However, recent findings have challenged this dogma and physiological levels of ROS are now considered as secondary messengers, mediating numerous cellular functions in stem cells. Stem cells represent important tools for tissue engineering, drug screening, and disease modeling. However, the safe use of stem cells for clinical applications still requires culture improvements to obtain functional cells. With the examples of mesenchymal stem cells (MSCs) and pluripotent stem cells (PSCs), this review investigates the roles of ROS in the maintenance of self-renewal, proliferation, and differentiation of stem cells. In addition, this work highlights that the tight control of stem cell microenvironment, including cell organization, and metabolic and mechanical environments, may be an effective approach to regulate endogenous ROS generation. Taken together, this paper indicates the need for better quantification of ROS towards the accurate control of stem cell fate.
Collapse
|
288
|
Klumpers DD, Mooney DJ, Smit TH. From Skeletal Development to Tissue Engineering: Lessons from the Micromass Assay. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:427-37. [PMID: 25946390 DOI: 10.1089/ten.teb.2014.0704] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Damage and degeneration of the skeletal elements due to disease, trauma, and aging lead to a significant health and economical burden. To reduce this burden, skeletal tissue engineering strategies aim to regenerate functional bone and cartilage in the adult body. However, challenges still exist. Such challenges involve the identification of the external cues that determine differentiation, how to control chondrocyte hypertrophy, and how to achieve specific tissue patterns and boundaries. To address these issues, it could be insightful to look at skeletal development, a robust morphogenetic process that takes place during embryonic development and is commonly modeled in vitro by the micromass assay. In this review, we investigate what the tissue engineering field can learn from this assay. By comparing embryonic skeletal precursor cells from different anatomic locations and developmental stages in micromass, the external cues that guide lineage commitment can be identified. The signaling pathways regulating chondrocyte hypertrophy, and the cues required for tissue patterning, can be elucidated by combining the micromass assay with genetic, molecular, and engineering tools. The lessons from the micromass assay are limited by two major differences between developmental and regenerative skeletogenesis: cell type and scale. We highlight an important difference between embryonic and adult skeletal progenitor cells, in that adult progenitors are not able to form mesenchymal condensations spontaneously. Also, the mechanisms of tissue patterning need to be adjusted to the larger tissue engineering constructs. In conclusion, mechanistic insights of skeletal tissue generation gained from the micromass model could lead to improved tissue engineering strategies and constructs.
Collapse
Affiliation(s)
- Darinka D Klumpers
- 1 School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts.,2 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,3 Department of Orthopedic Surgery, VU University Medical Centre MOVE Research Institute , Amsterdam, The Netherlands
| | - David J Mooney
- 1 School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts.,2 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Theo H Smit
- 3 Department of Orthopedic Surgery, VU University Medical Centre MOVE Research Institute , Amsterdam, The Netherlands
| |
Collapse
|
289
|
Qiu X, Zhang Y, Zhao X, Zhang S, Wu J, Guo H, Hu Y. Enhancement of endothelial differentiation of adipose derived mesenchymal stem cells by a three-dimensional culture system of microwell. Biomaterials 2015; 53:600-8. [DOI: 10.1016/j.biomaterials.2015.02.115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 12/15/2022]
|
290
|
Babur BK, Futrega K, Lott WB, Klein TJ, Cooper-White J, Doran MR. High-throughput bone and cartilage micropellet manufacture, followed by assembly of micropellets into biphasic osteochondral tissue. Cell Tissue Res 2015; 361:755-68. [PMID: 25924853 PMCID: PMC4550660 DOI: 10.1007/s00441-015-2159-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/22/2015] [Indexed: 11/05/2022]
Abstract
Engineered biphasic osteochondral tissues may have utility in cartilage defect repair. As bone-marrow-derived mesenchymal stem/stromal cells (MSC) have the capacity to make both bone-like and cartilage-like tissues, they are an ideal cell population for use in the manufacture of osteochondral tissues. Effective differentiation of MSC to bone-like and cartilage-like tissues requires two unique medium formulations and this presents a challenge both in achieving initial MSC differentiation and in maintaining tissue stability when the unified osteochondral tissue is subsequently cultured in a single medium formulation. In this proof-of-principle study, we used an in-house fabricated microwell platform to manufacture thousands of micropellets formed from 166 MSC each. We then characterized the development of bone-like and cartilage-like tissue formation in the micropellets maintained for 8–14 days in sequential combinations of osteogenic or chondrogenic induction medium. When bone-like or cartilage-like micropellets were induced for only 8 days, they displayed significant phenotypic changes when the osteogenic or chondrogenic induction medium, respectively, was swapped. Based on these data, we developed an extended 14-day protocol for the pre-culture of bone-like and cartilage-like micropellets in their respective induction medium. Unified osteochondral tissues were formed by layering 12,000 osteogenic micropellets and 12,000 chondrogenic micropellets into a biphasic structure and then further culture in chondrogenic induction medium. The assembled tissue was cultured for a further 8 days and characterized via histology. The micropellets had amalgamated into a continuous structure with distinctive bone-like and cartilage-like regions. This proof-of-concept study demonstrates the feasibility of micropellet assembly for the formation of osteochondral-like tissues for possible use in osteochondral defect repair.
Collapse
Affiliation(s)
- Betul Kul Babur
- Stem Cell Therapies Laboratory, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, Brisbane, Australia
| | | | | | | | | | | |
Collapse
|
291
|
The effect of growth-mimicking continuous strain on the early stages of skeletal development in micromass culture. PLoS One 2015; 10:e0124948. [PMID: 25915898 PMCID: PMC4411057 DOI: 10.1371/journal.pone.0124948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/19/2015] [Indexed: 11/19/2022] Open
Abstract
Embryonic skeletogenesis involves proliferation, condensation and subsequent chondrogenic differentiation of mesenchymal precursor cells, and the strains and stresses inherent to these processes have been hypothesized to influence skeletal development. The aim of this study was to determine the effect of growth-mimicking strain on the process of early skeletal development in vitro. To this end, we applied continuous uniaxial strain to embryonic skeletal precursor cells in micromass culture. Strain was applied at different times of culture to specifically address the effect of mechanical loading on the sequential stages of cellular proliferation, condensation and differentiation. We found that growth-mimicking strain at all three times did not affect proliferation or chondrogenic differentiation under the tested conditions. However, the timing of the applied strain did play a role in the density of mesenchymal condensations. This finding suggests that a mechanically dynamic environment, and specifically strain, can influence skeletal patterning. The growth-mimicking micromass model presented here may be a useful tool for further studies into the role of mechanical loading in early skeletal development.
Collapse
|
292
|
Kološa K, Motaln H, Herold-Mende C, Koršič M, Lah TT. Paracrine effects of mesenchymal stem cells induce senescence and differentiation of glioblastoma stem-like cells. Cell Transplant 2015; 24:631-44. [PMID: 25806680 DOI: 10.3727/096368915x687787] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma multiforme (GBM) displays high resistance to radiation and chemotherapy, due to the presence of a fraction of GBM stem-like cells (GSLCs), which are thus representing the target for GBM elimination. Since mesenchymal stem cells (MSCs) display high tumor tropism, we examined possible antitumor effects of the secreted factors from human MSCs on four GSLC lines (NCH421k, NCH644, NIB26, and NIB50). We found that conditioned media from bone marrow and umbilical cord-derived MSCs (MSC-CM) mediated cell cycle arrest of GSLCs by downregulating cyclin D1. PCR arrays revealed significantly deregulated expression of 13 genes associated with senescence in NCH421k cells exposed to MSC-CM. Among these, ATM, CD44, COL1A1, MORC3, NOX4, CDKN1A, IGFBP5, and SERPINE1 genes were upregulated, whereas IGFBP3, CDKN2A, CITED2, FN1, and PRKCD genes were found to be downregulated. Pathway analyses in GO and KEGG revealed their association with p53 signaling, which can trigger senescence via cell cycle inhibitors p21 or p16. For both, upregulated expression was proven in all four GSLC lines exhibiting senescence after MSC-CM exposure. Moreover, MSC paracrine signals were shown to increase the sensitivity of NCH421k and NCH644 cells toward temozolomide, possibly by altering them toward more differentiated cell types, as evidenced by vimentin and GFAP upregulation, and Sox-2 and Notch-1 downregulation. Our findings support the notion that MSCs posses an intrinsic ability to inhibit cell cycle and induce senescence and differentiation of GSLCs.
Collapse
Affiliation(s)
- Katja Kološa
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
293
|
Tsai AC, Liu Y, Yuan X, Ma T. Compaction, fusion, and functional activation of three-dimensional human mesenchymal stem cell aggregate. Tissue Eng Part A 2015; 21:1705-19. [PMID: 25661745 DOI: 10.1089/ten.tea.2014.0314] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are primary candidates in cell therapy and tissue engineering and are being tested in clinical trials for a wide range of diseases. Originally isolated and expanded as plastic adherent cells, hMSCs have intriguing properties of in vitro self-assembly into three-dimensional (3D) aggregates that improve a range of biological properties, including multilineage potential, secretion of therapeutic factors, and resistance against ischemic condition. While cell-cell contacts and cell-extracellular matrix interactions mediate 3D cell aggregation, the adaptive changes of hMSC cytoskeleton during self-assembly and associated metabolic reconfiguration may also influence aggregate properties and functional activation. In this study, we investigated the role of actin in regulating 3D hMSC aggregate compaction, fusion, spreading and functional activation. Individual hMSC aggregates with controlled initial cell number were formed by seeding a known number of hMSCs (500, 2000, and 5000 cells/well) in multi-well plates of an ultra-low adherent surface to form multicellular aggregates in individual wells. To assess the influence of actin-mediated contractility on hMSC aggregation and properties, actin modulators, including cytochalasin D (cytoD), nocodazole, lysophosphatidic acid (LPA), and Y-27632, were added at different stages of aggregation and their impacts on hMSC aggregate compaction and apoptosis were monitored. The results suggest that actin-mediated contractility influences hMSC aggregation, compaction, fusion, and spreading on adherent surface. Formation of multi-cellular aggregates significantly upregulated caspase 3/7 expression, expression of C-X-C chemokine receptor type 4 (CXCR-4), cell migration, secretion of prostaglandin E2 (PGE-2) and interleukin 6 (IL-6), and resistance to in vitro ischemic stress. The functional enhancement, however, is dependent on caspase activation, because treatment with Q-VD-OPh, a pan-caspase inhibitor, attenuated CXCR-4 and cytokine secretion. Importantly, comparable ATP/cell levels and significantly reduced mitochondrial membrane potential in aggregates of different sizes suggest that altered mitochondria bioenergetics on 3D aggregation is the primary inducer for apoptosis. Together, the results suggest multicellular aggregation as an effective and nongenetic strategy for hMSC functional activation.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, Florida State University , Tallahassee, Florida
| | | | | | | |
Collapse
|
294
|
Rajesh Krishnan G, Cheah C, Sarkar D. Hybrid Cross-Linking Characteristics of Hydrogel Control Stem Cell Fate. Macromol Biosci 2015; 15:747-55. [DOI: 10.1002/mabi.201400535] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 02/11/2015] [Indexed: 12/17/2022]
Affiliation(s)
- G. Rajesh Krishnan
- Department of Biomedical Engineering; University at Buffalo, The State University of New York; Buffalo NY 14260 USA
| | - Calvin Cheah
- Department of Biomedical Engineering; University at Buffalo, The State University of New York; Buffalo NY 14260 USA
| | - Debanjan Sarkar
- Department of Biomedical Engineering; University at Buffalo, The State University of New York; Buffalo NY 14260 USA
- Department of Chemical and Biological Engineering; University at Buffalo, The State University of New York; Buffalo NY 14260 USA
| |
Collapse
|
295
|
Lee J, Abdeen AA, Kim AS, Kilian KA. Influence of Biophysical Parameters on Maintaining the Mesenchymal Stem Cell Phenotype. ACS Biomater Sci Eng 2015; 1:218-226. [DOI: 10.1021/ab500003s] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Junmin Lee
- Department of Materials Science
and Engineering, Micro and Nanotechnology Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Amr A. Abdeen
- Department of Materials Science
and Engineering, Micro and Nanotechnology Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Alex S. Kim
- Department of Materials Science
and Engineering, Micro and Nanotechnology Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Kristopher A. Kilian
- Department of Materials Science
and Engineering, Micro and Nanotechnology Laboratory, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
296
|
Grayson WL, Bunnell BA, Martin E, Frazier T, Hung BP, Gimble JM. Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol 2015; 11:140-50. [PMID: 25560703 PMCID: PMC4338988 DOI: 10.1038/nrendo.2014.234] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem-cell-mediated bone repair has been used in clinical trials for the regeneration of large craniomaxillofacial defects, to slow the process of bone degeneration in patients with osteonecrosis of the femoral head and for prophylactic treatment of distal tibial fractures. Successful regenerative outcomes in these investigations have provided a solid foundation for wider use of stromal cells in skeletal repair therapy. However, employing stromal cells to facilitate or enhance bone repair is far from being adopted into clinical practice. Scientific, technical, practical and regulatory obstacles prevent the widespread therapeutic use of stromal cells. Ironically, one of the major challenges lies in the limited understanding of the mechanisms via which transplanted cells mediate regeneration. Animal models have been used to provide insight, but these models largely fail to reproduce the nuances of human diseases and bone defects. Consequently, the development of targeted approaches to optimize cell-mediated outcomes is difficult. In this Review, we highlight the successes and challenges reported in several clinical trials that involved the use of bone-marrow-derived mesenchymal or adipose-tissue-derived stromal cells. We identify several obstacles blocking the mainstream use of stromal cells to enhance skeletal repair and highlight technological innovations or areas in which novel techniques might be particularly fruitful in continuing to advance the field of skeletal regenerative medicine.
Collapse
Affiliation(s)
- Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Bruce A Bunnell
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Elizabeth Martin
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Trivia Frazier
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Ben P Hung
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Jeffrey M Gimble
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| |
Collapse
|
297
|
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
|
298
|
Wang Z, Xia J, Yan Y, Tsai AC, Li Y, Ma T, Guan J. Facile functionalization and assembly of live cells with microcontact-printed polymeric biomaterials. Acta Biomater 2015; 11:80-7. [PMID: 25305514 DOI: 10.1016/j.actbio.2014.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/23/2014] [Accepted: 10/02/2014] [Indexed: 01/11/2023]
Abstract
The functionalization and assembly of live cells with microfabricated polymeric biomaterials have attracted considerable interest in recent years, but the conventional methods suffer from high cost, high complexity, long processing time or inadequate capability. The present study reports on the development of a novel method for functionalizing and assembling live cells by integrating microcontact printing of polymeric biomaterials with a temperature-sensitive sacrificial layer prepared by spin-coating. This method has been used not only to functionalize live cells with microscopic polyelectrolyte and thermoplastic structures of various sizes and shapes, but also to assemble the cells into macroscopic stripes and sheets. The method is applicable to multiple types of cells, including human leukemic cells, mouse embryonic stem cells and human mesenchymal stem cells in the forms of single cells and cell aggregates. In addition, the microcontact-printed structures can be prepared using biodegradable and biocompatible polyelectrolytes and thermoplastic. The unique combination of low cost, ease of use and high versatility renders this method potentially useful for diverse biomedical applications, including drug delivery, cell tracking and tissue engineering.
Collapse
|
299
|
Bogdanova-Jatniece A, Berzins U, Kozlovska T. Growth Properties and Pluripotency Marker Expression of Spontaneously Formed Three-dimensional Aggregates of Human Adipose-derived Stem Cells. Int J Stem Cells 2014; 7:143-52. [PMID: 25473452 PMCID: PMC4249897 DOI: 10.15283/ijsc.2014.7.2.143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2014] [Indexed: 12/28/2022] Open
Abstract
Background and Objectives: Recent findings suggest that therapeutic potential of mesenchymal stem cells (MSCs) could be increased through aggregation into three-dimensional (3D) bodies, and different culture methods have been employed to obtain 3D spheroids of MSCs. In the current study we report accidentally encountered spontaneous formation of adipose-derived stem cell (ASC) bodies in standard ASC culture of a single donor. Methods and Results: Human ASCs from passages 1 to 3, cultured in a medium containing 5% autologous serum (AS), spontaneously clustered and formed floating 3D bodies. After a transfer of floating ASC bodies onto new adherent plastic dish, they attached to the surface and gradual migration of spindle-shaped ASCs out of the bodies was detected. A substitution of AS with allogeneic sera did not hinder this ability, but commercial medium containing fetal bovine serum delayed the process. Substantial part of ASCs surrounding transferred ASC bodies showed alkaline phosphatase (AP) activity, while ASC aggregates were AP negative. Similar 3D bodies formed when ASCs were grown on an uncoated glass surface. These ASC aggregates as well as clusters of ASCs, where formation of the 3D bodies is initiated, expressed pluripotency marker NANOG, but the expression of OCT4A was not detected. Conclusions: Obtained results suggest that spontaneously formed ASC aggregates may represent a more primitive cell subpopulation within the individual ASC culture. The ability to form 3D aggregates, the expression of NANOG, and the lack of the AP activity may be used to enrich ASC cultures with potentially more primitive cells serving as an excellent basis for therapeutic applications.
Collapse
Affiliation(s)
| | - Uldis Berzins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | |
Collapse
|
300
|
Chien HW, Fu SW, Shih AY, Tsai WB. Modulation of the stemness and osteogenic differentiation of human mesenchymal stem cells by controlling RGD concentrations of poly(carboxybetaine) hydrogel. Biotechnol J 2014; 9:1613-23. [PMID: 25303097 DOI: 10.1002/biot.201300433] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 08/24/2014] [Accepted: 10/08/2014] [Indexed: 12/15/2022]
Abstract
In vitro modulation of the differentiation status of mesenchymal stem cells (MSCs) is important for their application to regenerative medicine. We suggested that the morphology and differentiation states of MSCs could be modulated by controlling the cell affinity of a substrate. The objective of this study was to investigate the effects of surface bio-adhesive signals on self-renewal and osteogenic differentiation of MSCs using a low-fouling platform. Cell-resistant poly(carboxybetaine) hydrogel was conjugated with 5 μM or 5 mM of cell-adhesive arginine-glycine-aspartic acid (RGD) peptides in order to control the cells' affinity to the substrate. Human mesenchymal stem cells (hMSCs) were cultured on the RGD-modified poly(carboxybetaine) hydrogel and then the cells' states of stemness and osteogenic differentiation were evaluated using reverse-transcriptase polymerase chain reaction. The hMSCs formed three-dimensional spheroids on the 5 μM RGD substrate, while cells on the 5 mM RGD substrate exhibited spreading morphology. Furthermore, cells on the 5 μM RGD hydrogel maintained a better stemness phenotype, while the hMSCs on the 5 mM RGD hydrogel proliferated faster and underwent osteogenic differentiation. In conclusion, the stemness of hMSCs was best maintained on a low RGD surface, while osteogenic differentiation of hMSCs was enhanced on a high RGD surface.
Collapse
Affiliation(s)
- Hsiu-Wen Chien
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|