1
|
Zhang Z, Gong N, Wang Y, Xu L, Zhao S, Liu Y, Tan F. Impact of Strontium, Magnesium, and Zinc Ions on the In Vitro Osteogenesis of Maxillary Sinus Membrane Stem Cells. Biol Trace Elem Res 2024:10.1007/s12011-024-04303-4. [PMID: 39150638 DOI: 10.1007/s12011-024-04303-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/03/2024] [Indexed: 08/17/2024]
Abstract
Human Maxillary Sinus Membrane Stem Cells (hMSMSCs) contribute significantly to bone formation following maxillary sinus floor augmentation (MSFA). The biological behavior of mesenchymal stem cells is notably influenced by varying concentrations of magnesium (Mg2+), strontium (Sr2+), and zinc (Zn2+) ions; however, their specific effects on hMSMSCs have not been comprehensively studied. We isolated hMSMSCs and identified their mesenchymal stem cell characteristics by flow cytometry and multilineage differentiation experiments. Subsequently, the hMSMSCs were cultured in media containing different concentrations of these metal ions. The proliferation and viability of hMSMSCs were assessed using CCK-8 and Calcein AM/PI staining. After osteogenic induction, cells were evaluated for alkaline phosphatase (ALP) activity, ALP staining, and Alizarin Red staining. Additionally, qRT-PCR was used to detect differences in osteogenic gene expression, and immunofluorescence staining was used to observe variations in OCN protein levels. The results indicated that 1 mM Mg2+, 0.01 mM Sr2+, and 0.001 mM Zn2+ significantly improved the proliferation and activity of hMSMSCs. These concentrations also notably enhanced ALP secretion, increased bone-related gene expression, and augmented osteocalcin expression and formation of extracellular calcium nodules, thereby improving osteogenic differentiation. However, higher concentrations of Mg2+, Sr2+, and Zn2+ decreased cell viability and osteogenic differentiation. Mg2+, Sr2+, and Zn2+ promote osteogenic differentiation and proliferation of hMSMSCs in a concentration-dependent manner, indicating that the type and concentration of ions in the extracellular environment can significantly alter hMSMSCs behavior, which is a crucial consideration for material design in maxillary sinus elevation applications.
Collapse
Affiliation(s)
- Zhihao Zhang
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Ning Gong
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Ying Wang
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Lei Xu
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Sinan Zhao
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yanshan Liu
- School of Stomatology, Qingdao University, Qingdao, 266023, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Fei Tan
- Department of Prosthodontic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- School of Stomatology, Qingdao University, Qingdao, 266023, China.
| |
Collapse
|
2
|
Hanetseder D, Levstek T, Teuschl-Woller AH, Frank JK, Schaedl B, Redl H, Marolt Presen D. Engineering of extracellular matrix from human iPSC-mesenchymal progenitors to enhance osteogenic capacity of human bone marrow stromal cells independent of their age. Front Bioeng Biotechnol 2023; 11:1214019. [PMID: 37600321 PMCID: PMC10434254 DOI: 10.3389/fbioe.2023.1214019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Regeneration of bone defects is often limited due to compromised bone tissue physiology. Previous studies suggest that engineered extracellular matrices enhance the regenerative capacity of mesenchymal stromal cells. In this study, we used human-induced pluripotent stem cells, a scalable source of young mesenchymal progenitors (hiPSC-MPs), to generate extracellular matrix (iECM) and test its effects on the osteogenic capacity of human bone-marrow mesenchymal stromal cells (BMSCs). iECM was deposited as a layer on cell culture dishes and into three-dimensional (3D) silk-based spongy scaffolds. After decellularization, iECM maintained inherent structural proteins including collagens, fibronectin and laminin, and contained minimal residual DNA. Young adult and aged BMSCs cultured on the iECM layer in osteogenic medium exhibited a significant increase in proliferation, osteogenic marker expression, and mineralization as compared to tissue culture plastic. With BMSCs from aged donors, matrix mineralization was only detected when cultured on iECM, but not on tissue culture plastic. When cultured in 3D iECM/silk scaffolds, BMSCs exhibited significantly increased osteogenic gene expression levels and bone matrix deposition. iECM layer showed a similar enhancement of aged BMSC proliferation, osteogenic gene expression, and mineralization compared with extracellular matrix layers derived from young adult or aged BMSCs. However, iECM increased osteogenic differentiation and decreased adipocyte formation compared with single protein substrates including collagen and fibronectin. Together, our data suggest that the microenvironment comprised of iECM can enhance the osteogenic activity of BMSCs, providing a bioactive and scalable biomaterial strategy for enhancing bone regeneration in patients with delayed or failed bone healing.
Collapse
Affiliation(s)
- Dominik Hanetseder
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Tina Levstek
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Herbert Teuschl-Woller
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Julia Katharina Frank
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
3
|
Bloise N, Waldorff EI, Montagna G, Bruni G, Fassina L, Fang S, Zhang N, Jiang J, Ryaby JT, Visai L. Early Osteogenic Marker Expression in hMSCs Cultured onto Acid Etching-Derived Micro- and Nanotopography 3D-Printed Titanium Surfaces. Int J Mol Sci 2022; 23:7070. [PMID: 35806083 PMCID: PMC9266831 DOI: 10.3390/ijms23137070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022] Open
Abstract
Polyetheretherketone (PEEK) titanium composite (PTC) is a novel interbody fusion device that combines a PEEK core with titanium alloy (Ti6Al4V) endplates. The present study aimed to investigate the in vitro biological reactivity of human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) to micro- and nanotopographies produced by an acid-etching process on the surface of 3D-printed PTC endplates. Optical profilometer and scanning electron microscopy were used to assess the surface roughness and identify the nano-features of etched or unetched PTC endplates, respectively. The viability, morphology and the expression of specific osteogenic markers were examined after 7 days of culture in the seeded cells. Haralick texture analysis was carried out on the unseeded endplates to correlate surface texture features to the biological data. The acid-etching process modified the surface roughness of the 3D-printed PTC endplates, creating micro- and nano-scale structures that significantly contributed to sustaining the viability of hBM-MSCs and triggering the expression of early osteogenic markers, such as alkaline phosphatase activity and bone-ECM protein production. Finally, the topography of 3D-printed PTC endplates influenced Haralick's features, which in turn correlated with the expression of two osteogenic markers, osteopontin and osteocalcin. Overall, these data demonstrate that the acid-etching process of PTC endplates created a favourable environment for osteogenic differentiation of hBM-MSCs and may potentially have clinical benefit.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| | - Erik I. Waldorff
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Giulia Montagna
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Giovanna Bruni
- C.S.G.I.-Department of Chemistry, Section of Physical Chemistry, University of Pavia, 27100 Pavia, Italy;
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Samuel Fang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Nianli Zhang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Jiechao Jiang
- Department of Material Science, University of Texas, Arlington, TX 76019, USA;
| | - James T. Ryaby
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Livia Visai
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| |
Collapse
|
4
|
Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal Stromal Cell-Based Bone Regeneration Therapies: From Cell Transplantation and Tissue Engineering to Therapeutic Secretomes and Extracellular Vesicles. Front Bioeng Biotechnol 2019; 7:352. [PMID: 31828066 PMCID: PMC6890555 DOI: 10.3389/fbioe.2019.00352] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Effective regeneration of bone defects often presents significant challenges, particularly in patients with decreased tissue regeneration capacity due to extensive trauma, disease, and/or advanced age. A number of studies have focused on enhancing bone regeneration by applying mesenchymal stromal cells (MSCs) or MSC-based bone tissue engineering strategies. However, translation of these approaches from basic research findings to clinical use has been hampered by the limited understanding of MSC therapeutic actions and complexities, as well as costs related to the manufacturing, regulatory approval, and clinical use of living cells and engineered tissues. More recently, a shift from the view of MSCs directly contributing to tissue regeneration toward appreciating MSCs as "cell factories" that secrete a variety of bioactive molecules and extracellular vesicles with trophic and immunomodulatory activities has steered research into new MSC-based, "cell-free" therapeutic modalities. The current review recapitulates recent developments, challenges, and future perspectives of these various MSC-based bone tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Traweger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Spinal Cord Injury & Tissue Regeneration Center Salzburg, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
5
|
Chai L, Quan R, Hu J, Huang X, Lü J, Zhang C, Qiu R, Cai B. [ In vitro study of bone morphogenetic protein 2 gelatin/chitosan hydrogel sustained-release system composite hydroxyapatite/zirconium dioxide foam ceramics and induced pluripotent stem cells derived mesenchymal stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:252-258. [PMID: 30739425 PMCID: PMC8337614 DOI: 10.7507/1002-1892.201809060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/10/2019] [Indexed: 11/03/2022]
Abstract
Objective To construct bone morphogenetic protein 2 (BMP-2) gelatin/chitosan hydrogel sustained-release system, co-implant with induced pluripotent stem cells (iPS) derived mesenchymal stem cells (MSCs) to hydroxyapatite (HA)/zirconium dioxide (ZrO 2) bio porous ceramic foam, co-culture in vitro, and to explore the effect of sustained-release system on osteogenic differentiation of iPS-MSCs. Methods BMP-2 gelatin/chitosan hydrogel microspheres were prepared by water-in-oil solution. Drug encapsulation efficiency, drug loading, and in vitro sustained release rate of the microspheres were tested. HA/ZrO 2 bio porous ceramic foam composite iPS-MSCs and BMP-2 gelatin/chitosan hydrogel sustained release system co-culture system was established as experimental group, and cell scaffold complex without BMP-2 composite gelatin/chitosan hydrogel sustained release system as control group. After 3, 7, 10, and 14 days of co-culture in the two groups, ALP secretion of cells was detected; gene expression levels of core binding factor alpha 1 (Cbfa1), collagen type Ⅰ, and Osterix (OSX) were detected by RT-PCR; the expression of collagen type Ⅰ was observed by immunohistochemical staining at 14 days of culture; and cell creep and adhesion were observed by scanning electron microscopy. Results BMP-2 gelatin/chitosan hydrogel sustained-release system had better drug encapsulation efficiency and drug loading, and could prolong the activity time of BMP-2. The secretion of ALP and the relative expression of Cbfa1, collagen type Ⅰ, and OSX genes in the experimental group were significantly higher than those in the control group at different time points in the in vitro co-culture system ( P<0.05). Immunohistochemical staining showed that the amount of fluorescence in the experimental group was significantly more than that in the control group, i.e. the expression level of collagen type Ⅰ was higher than that in the control group. The cells could be more evenly distributed on the materials, and the cell morphology was good. Scanning electron microscopy showed that the sustained-release system could adhere to cells well. Conclusion iPS-MSCs have the ability of osteogenic differentiation, which is significantly enhanced by BMP-2 gelatin/chitosan hydrogel sustained-release system. The combination of iPS-MSCs and sustained-release system can adhere to the materials well, and the cell activity is better.
Collapse
Affiliation(s)
- Le Chai
- Zhejiang University of Traditional Chinese Medicine, Hangzhou Zhejiang, 310053, P.R.China
| | - Renfu Quan
- Department of Spine Surgery, Jiangnan Hospital of Zhejiang Chinese Medicine College, Hangzhou Zhejiang, 311200,
| | - Jintao Hu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou Zhejiang, 310053, P.R.China
| | - Xiaolong Huang
- Department of Spine Surgery, Jiangnan Hospital of Zhejiang Chinese Medicine College, Hangzhou Zhejiang, 311200, P.R.China
| | - Jianlan Lü
- Zhejiang University of Traditional Chinese Medicine, Hangzhou Zhejiang, 310053, P.R.China
| | - Can Zhang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou Zhejiang, 310053, P.R.China
| | - Rui Qiu
- Department of Spine Surgery, Jiangnan Hospital of Zhejiang Chinese Medicine College, Hangzhou Zhejiang, 311200, P.R.China
| | - Bingbing Cai
- Department of Spine Surgery, Jiangnan Hospital of Zhejiang Chinese Medicine College, Hangzhou Zhejiang, 311200, P.R.China
| |
Collapse
|
6
|
Ort C, Dayekh K, Xing M, Mequanint K. Emerging Strategies for Stem Cell Lineage Commitment in Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2018; 4:3644-3657. [PMID: 33429592 DOI: 10.1021/acsbiomaterials.8b00532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem cells have transformed the fields of tissue engineering and regenerative medicine, and their potential to further advance these fields cannot be overstated. The stem cell niche is a dynamic microenvironment that determines cell fate during development and tissue repair following an injury. Classically, stem cells were studied in isolation of their microenvironment; however, contemporary research has produced a myriad of evidence that shows the importance of multiple aspects of the stem cell niche in regulating their processes. In the context of tissue engineering and regenerative medicine studies, the niche is an artificial environment provided by culture conditions. In vitro culture conditions may involve coculturing with other cell types, developing specific biomaterials, and applying relevant forces to promote the desired lineage commitment. Considerable advance has been made over the past few years toward directed stem cell differentiation; however, the unspecific differentiation of stem cells yielding a mixed population of cells has been a challenge. In this review, we provide a systematic review of the emerging strategies used for lineage commitment within the context of tissue engineering and regenerative medicine. These strategies include scaffold pore-size and pore-shape gradients, stress relaxation, sonic and electromagnetic effects, and magnetic forces. Finally, we provide insights and perspectives into future directions focusing on signaling pathways activated during lineage commitment using external stimuli.
Collapse
Affiliation(s)
| | | | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, 66 Chancellors Circle, Winnipeg R3T 2N2, Canada
| | | |
Collapse
|
7
|
Luzzani CD, Miriuka SG. Pluripotent Stem Cells as a Robust Source of Mesenchymal Stem Cells. Stem Cell Rev Rep 2017; 13:68-78. [PMID: 27815690 DOI: 10.1007/s12015-016-9695-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSC) have been extensively studied over the past years for the treatment of different diseases. Most of the ongoing clinical trials currently involve the use of MSC derived from adult tissues. This source may have some limitations, particularly with therapies that may require extensive and repetitive cell dosage. However, nowadays, there is a staggering growth in literature on a new source of MSC. There is now increasing evidence about the mesenchymal differentiation from pluripotent stem cell (PSC). Here, we summarize the current knowledge of pluripotent-derived mesenchymal stem cells (PD-MSC). We present a historical perspective on the subject, and then discuss some critical questions that remain unanswered.
Collapse
Affiliation(s)
- Carlos D Luzzani
- LIAN-CONICET - FLENI, Ruta 9 Km 52, 5 - (B1625XAF) Belén de Escobar, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Santiago G Miriuka
- LIAN-CONICET - FLENI, Ruta 9 Km 52, 5 - (B1625XAF) Belén de Escobar, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
8
|
Characterization of Mesenchymal Stem Cell-Like Cells Derived From Human iPSCs via Neural Crest Development and Their Application for Osteochondral Repair. Stem Cells Int 2017; 2017:1960965. [PMID: 28607560 PMCID: PMC5451770 DOI: 10.1155/2017/1960965] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/03/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from induced pluripotent stem cells (iPSCs) are a promising cell source for the repair of skeletal disorders. Recently, neural crest cells (NCCs) were reported to be effective for inducing mesenchymal progenitors, which have potential to differentiate into osteochondral lineages. Our aim was to investigate the feasibility of MSC-like cells originated from iPSCs via NCCs for osteochondral repair. Initially, MSC-like cells derived from iPSC-NCCs (iNCCs) were generated and characterized in vitro. These iNCC-derived MSC-like cells (iNCMSCs) exhibited a homogenous population and potential for osteochondral differentiation. No upregulation of pluripotent markers was detected during culture. Second, we implanted iNCMSC-derived tissue-engineered constructs into rat osteochondral defects without any preinduction for specific differentiation lineages. The implanted cells remained alive at the implanted site, whereas they failed to repair the defects, with only scarce development of osteochondral tissue in vivo. With regard to tumorigenesis, the implanted cells gradually disappeared and no malignant cells were detected throughout the 2-month follow-up. While this study did not show that iNCMSCs have efficacy for repair of osteochondral defects when implanted under undifferentiated conditions, iNCMSCs exhibited good chondrogenic potential in vitro under appropriate conditions. With further optimization, iNCMSCs may be a new source for tissue engineering of cartilage.
Collapse
|
9
|
Maisani M, Pezzoli D, Chassande O, Mantovani D. Cellularizing hydrogel-based scaffolds to repair bone tissue: How to create a physiologically relevant micro-environment? J Tissue Eng 2017; 8:2041731417712073. [PMID: 28634532 PMCID: PMC5467968 DOI: 10.1177/2041731417712073] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022] Open
Abstract
Tissue engineering is a promising alternative to autografts or allografts for the regeneration of large bone defects. Cell-free biomaterials with different degrees of sophistication can be used for several therapeutic indications, to stimulate bone repair by the host tissue. However, when osteoprogenitors are not available in the damaged tissue, exogenous cells with an osteoblast differentiation potential must be provided. These cells should have the capacity to colonize the defect and to participate in the building of new bone tissue. To achieve this goal, cells must survive, remain in the defect site, eventually proliferate, and differentiate into mature osteoblasts. A critical issue for these engrafted cells is to be fed by oxygen and nutrients: the transient absence of a vascular network upon implantation is a major challenge for cells to survive in the site of implantation, and different strategies can be followed to promote cell survival under poor oxygen and nutrient supply and to promote rapid vascularization of the defect area. These strategies involve the use of scaffolds designed to create the appropriate micro-environment for cells to survive, proliferate, and differentiate in vitro and in vivo. Hydrogels are an eclectic class of materials that can be easily cellularized and provide effective, minimally invasive approaches to fill bone defects and favor bone tissue regeneration. Furthermore, by playing on their composition and processing, it is possible to obtain biocompatible systems with adequate chemical, biological, and mechanical properties. However, only a good combination of scaffold and cells, possibly with the aid of incorporated growth factors, can lead to successful results in bone regeneration. This review presents the strategies used to design cellularized hydrogel-based systems for bone regeneration, identifying the key parameters of the many different micro-environments created within hydrogels.
Collapse
Affiliation(s)
- Mathieu Maisani
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
- Laboratoire BioTis, Inserm U1026, Université de Bordeaux, Bordeaux, France
| | - Daniele Pezzoli
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
| | - Olivier Chassande
- Laboratoire BioTis, Inserm U1026, Université de Bordeaux, Bordeaux, France
| | - Diego Mantovani
- Laboratory for Biomaterials & Bioengineering (CRC-I), Department Min-Met-Materials Engineering & Research Center CHU de Québec, Laval University, Québec City, QC, Canada
| |
Collapse
|
10
|
Ding S, Kingshott P, Thissen H, Pera M, Wang PY. Modulation of human mesenchymal and pluripotent stem cell behavior using biophysical and biochemical cues: A review. Biotechnol Bioeng 2016; 114:260-280. [DOI: 10.1002/bit.26075] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/27/2016] [Accepted: 08/07/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Sheryl Ding
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
| | - Peter Kingshott
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
| | | | - Martin Pera
- Department of Anatomy and Neuroscience, Walter and Eliza Hall Institute of Medical Research, Florey Neuroscience and Mental Health Institute; The University of Melbourne; Victoria Australia
| | - Peng-Yuan Wang
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
- CSIRO Manufacturing; Clayton Victoria Australia
- Department of Anatomy and Neuroscience, Walter and Eliza Hall Institute of Medical Research, Florey Neuroscience and Mental Health Institute; The University of Melbourne; Victoria Australia
- Graduate Institute of Nanomedicine and Medical Engineering; College of Biomedical Engineering; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
11
|
Li S, Hu C, Li J, Liu L, Jing W, Tang W, Tian W, Long J. Effect of miR-26a-5p on the Wnt/Ca(2+) Pathway and Osteogenic Differentiation of Mouse Adipose-Derived Mesenchymal Stem Cells. Calcif Tissue Int 2016; 99:174-86. [PMID: 27040676 DOI: 10.1007/s00223-016-0137-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/24/2016] [Indexed: 12/31/2022]
Abstract
Elucidation of the molecular mechanisms that regulate the differentiation of adipose-derived mesenchymal stem cells into osteogenic cells may lead to new methods for bone tissue engineering. We examined the role of miR-26a-5p in the regulation of osteogenic differentiation of mouse adipose-derived mesenchymal stem cells (mADSCs) by using mimics and inhibitors of this microRNA. Our results showed that over-expression of miR-26a-5p inhibited osteogenesis and that suppression of endogenous miR-26a-5p promoted osteogenesis. Four bioinformatics algorithms indicated that the 3'UTR of Wnt5a was a potential target of miR-26a-5p. We confirmed this prediction by use of dual-luciferase reporter assay and GFP/RFP assay. We also examined the molecular mechanisms by which miR-26a-5p regulates osteogenesis. Fura-2AM and Western blot assays after transfection indicated that miR-26a-5p repressed WNT5A, inhibited calcium flux and protein kinase C, and suppressed osteogenic differentiation of mADSCs. By contrast, miR-26a-5p inhibition activated these signal proteins and promoted osteogenic differentiation. Taken together, our results suggest that up-regulation of miR-26a-5p inhibits osteogenic differentiation of mADSCs by directly targeting the 3'UTR of Wnt5a, thereby down-regulating the Wnt/Ca(2+) signaling pathway.
Collapse
Affiliation(s)
- Shasha Li
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Chen Hu
- Department of Oral and Maxillofacial Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Jianwei Li
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Lei Liu
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Wei Jing
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Wei Tang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Weidong Tian
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
12
|
Biomechanics: Principles. Bioengineering (Basel) 2015. [DOI: 10.1007/978-3-319-10798-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
13
|
|
14
|
|
15
|
Cultivation of human bone-like tissue from pluripotent stem cell-derived osteogenic progenitors in perfusion bioreactors. Methods Mol Biol 2013; 1202:173-84. [PMID: 24281874 DOI: 10.1007/7651_2013_52] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human pluripotent stem cells represent an unlimited source of skeletal tissue progenitors for studies of bone biology, pathogenesis, and the development of new approaches for bone reconstruction and therapies. In order to construct in vitro models of bone tissue development and to grow functional, clinical-size bone substitutes for transplantation, cell cultivation in three-dimensional environments composed of porous osteoconductive scaffolds and dynamic culture systems-bioreactors-has been studied. Here, we describe a stepwise procedure for the induction of human embryonic and induced pluripotent stem cells (collectively termed PSCs) into mesenchymal-like progenitors, and their subsequent cultivation on decellularized bovine bone scaffolds in perfusion bioreactors, to support the development of viable, stable bone-like tissue in defined geometries.
Collapse
|