1
|
Zhang C, Qian C, Yang G, Zhu Y, Kang B, Chen X, Chen S. Microarc Oxidation Coatings Doped with a Low Proportion of Yttrium Enhance the Osseointegration of Titanium Implants through the BMP/Smad Pathway. ACS Biomater Sci Eng 2025; 11:1869-1881. [PMID: 39945293 PMCID: PMC11897939 DOI: 10.1021/acsbiomaterials.4c02461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 03/11/2025]
Abstract
Adding metal ions is a promising strategy to enhance the biological performance of titanium implants. In this study, we aimed to explore the effects of yttrium on the osseointegration of titanium implants. First, a series of yttrium-doped titanium surfaces were fabricated via microarc oxidation (MAO) by incorporating yttrium acetate into the electrolyte, and then the surface characteristics of different substrates were evaluated. Subsequently, the cellular behaviors of different coatings were assessed, and the osteointegration effects were examined using a rat model. Finally, high-throughput sequencing was employed to elucidate the underlying mechanisms of the yttrium-doped MAO coatings. As the results indicated, the proportion of yttrium in the coatings increased as the concentration of yttrium acetate improved. Surface characterization revealed that the yttrium-doped MAO coatings exhibited a homogeneous porous morphology, with comparable roughness and wettability to those of the undoped MAO coating, while the morphology became inconsistent when the yttrium acetate concentration reached 30 mM. The in vitro assays demonstrated that the addition of yttrium notably improved the cell adhesion, spreading, proliferation, and osteogenic differentiation of MAO coatings when doped with a low proportion, accompanied by enhanced osseointegration according to the in vivo experiments. Further exploration revealed a significant enrichment of osseointegration-related signaling factors and the activation of BMP/Smad signaling in the effects of yttrium-doped titanium coatings, which was attributed to the excessive accumulation of phosphorylated Smad1/5/9 in the nucleus. In summary, our work demonstrates that the use of MAO coatings doped with a low proportion of yttrium can enhance the osseointegration of titanium implants, providing an efficient strategy to optimize titanium implant performance.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department
of Oral Implantology, Shanghai Key Laboratory of Craniomaxillofacial
Development and Diseases, Shanghai Stomatological Hospital & School
of Stomatology, Fudan University, Shanghai 200001, China
| | - Chenghui Qian
- Department
of Multidisciplinary Consultant Center, Shanghai Key Laboratory of
Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan
University, Shanghai 200001, China
| | - Guang Yang
- School
of Materials and Chemistry, University of
Shanghai for Science and Technology, Shanghai 200093, China
| | - Yiying Zhu
- Department
of Oral Implantology, Shanghai Key Laboratory of Craniomaxillofacial
Development and Diseases, Shanghai Stomatological Hospital & School
of Stomatology, Fudan University, Shanghai 200001, China
| | - Binbin Kang
- School
of Materials and Chemistry, University of
Shanghai for Science and Technology, Shanghai 200093, China
| | - Xiaohong Chen
- School
of Materials and Chemistry, University of
Shanghai for Science and Technology, Shanghai 200093, China
| | - Si Chen
- Department
of Multidisciplinary Consultant Center, Shanghai Key Laboratory of
Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan
University, Shanghai 200001, China
| |
Collapse
|
2
|
Gaihre B, Camilleri E, Tilton M, Astudillo Potes MD, Liu X, Lucien F, Lu L. LAPONITE® nano-silicates potentiate the angiogenic effects of FG-4592 and osteogenic effects of BMP-2. Biomater Sci 2024; 12:5610-5619. [PMID: 39359127 PMCID: PMC11822916 DOI: 10.1039/d4bm00636d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
LAPONITE®-based drug delivery systems offer many advantages due to the unique ionic and physical properties of LAPONITE®. The high ionicity and large surface area of LAPONITE® nanoparticles enable the intercalation and dissolution of biomolecules. In this study, we explored the potential of LAPONITE® as a carrier for FG-4592 to support angiogenesis and as a carrier for bone morphogenic protein-2 (BMP-2) to support osteogenesis. Interestingly, we found that LAPONITE® promoted the FG-4592 induced upregulation of vascular endothelial growth factor (VEGF) gene expression of human umbilical cord endothelial cells (HUVECs). Additionally, we observed that LAPONITE® could provide a sustained release of BMP-2 and significantly potentiate the osteogenic effects of BMP-2 on adipose derived mesenchymal stem cells (AMSCs). Overall, current findings on the LAPONITE®-drug/protein model system provide a unique way to potentiate the angiogenic activities of FG-4592 on HUVECs and osteogenic effects of BMP-2 on AMSCs for tissue engineering application. Future studies will be directed towards gaining a deeper understanding of these effects on a co-culture system of HUVECs and AMSCs.
Collapse
Affiliation(s)
- Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emily Camilleri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maria D Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
3
|
Weston JD, Austin B, Levis H, Zitnay J, Weiss JA, Lawrence B, Bowles RD. Toward the Development of a Tissue Engineered Gradient Utilizing CRISPR-Guided Gene Modulation. Tissue Eng Part A 2024; 30:525-535. [PMID: 38323556 DOI: 10.1089/ten.tea.2023.0352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024] Open
Abstract
Cellular, compositional, and mechanical gradients are found throughout biological tissues, especially in transition zones between tissue types. Yet, strategies to engineer such gradients have proven difficult due to the complex nature of these tissues. Current strategies for tissue engineering complex gradients often utilize stem cells; however, these multipotent cells require direction from environmental cues, which can be difficult to control both in vitro and in vivo. In this study, we utilize clustered regularly-interspaced short palindromic repeats (CRISPR)-guided gene modulation to direct the differentiation of multipotent adipose-derived stem cells (ASCs) to demonstrate the effectiveness of CRISPR-engineered cells in tissue engineering applications. Specifically, we screen CRISPR-interference (CRISPRi) constructs targeting the promotors of selected osteogenic inhibitors and demonstrate that ASC osteogenic differentiation and mineral deposition can be regulated with CRISPRi targeting of Noggin without the use of exogenous growth factors in tissue engineered constructs. As a proof of concept, we combine three technologies developed out of our laboratories to demonstrate the controlled deposition of these engineered cells in a gradient with CRISPR-activation multiplex-engineered aggrecan/collagen type-II-chondrogenic ASCs on a high density anisotropic type I collagen construct to create a cell and tissue gradient similar to the fibrocartilage-to-mineralized-fibrocartilage gradient in the enthesis. Our results display the promise of CRISPR-engineered ASCs to produce tissue gradients, similar to what is observed in native tissue.
Collapse
Affiliation(s)
- Jacob D Weston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Brooke Austin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Hunter Levis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jared Zitnay
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Brandon Lawrence
- Department of Orthopedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Robby D Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
5
|
Çakmak A, Fuerkaiti S, Karagüzel D, Karaaslan Ç, Gümüşderelioğlu M. Enhanced Osteogenic Potential of Noggin Knockout C2C12 Cells on BMP-2 Releasing Silk Scaffolds. ACS Biomater Sci Eng 2023; 9:6175-6185. [PMID: 37796024 PMCID: PMC10646847 DOI: 10.1021/acsbiomaterials.3c00506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023]
Abstract
The CRISPR/Cas9 mechanism offers promising therapeutic approaches for bone regeneration by stimulating or suppressing critical signaling pathways. In this study, we aimed to increase the activity of BMP-2 signaling through knockout of Noggin, thereby establishing a synergistic effect on the osteogenic activity of cells in the presence of BMP-2. Since Noggin is an antagonist expressed in skeletal tissues and binds to subunits of bone morphogenetic proteins (BMPs) to inhibit osteogenic differentiation, here Noggin expression was knocked out using the CRISPR/Cas9 system. In accordance with this purpose, C2C12 (mouse myoblast) cells were transfected with CRISPR/Cas9 plasmids. Transfection was achieved with Lipofectamine and confirmed with intense fluorescent signals in microscopic images and deletion in target sequence in Sanger sequencing analysis. Thus, Noggin knockout cells were identified as a new cell source for tissue engineering studies. Then, the transfected cells were seeded on highly porous silk scaffolds bearing BMP-2-loaded silk nanoparticles (30 ng BMP-2/mg silk nanoparticle) in the size of 288 ± 62 nm. BMP-2 is released from the scaffolds in a controlled manner for up to 60 days. The knockout of Noggin by CRISPR/Cas9 was found to synergistically promote osteogenic differentiation in the presence of BMP-2 through increased Coll1A1 and Ocn expression and mineralization. Gene editing of Noggin and BMP-2 increased almost 2-fold Col1A1 expression and almost 3-fold Ocn expression compared to the control group. Moreover, transfected cells produced extracellular matrix (ECM) containing collagen fibers on the scaffolds and mineral-like structures were formed on the fibers. In addition, mineralization characterized by intense Alizarin red staining was detected in transfected cells cultured in the presence of BMP-2, while the other groups did not exhibit any mineralized areas. As has been demonstrated in this study, the CRISPR/Cas9 mechanism has great potential for obtaining new cell sources to be used in tissue engineering studies.
Collapse
Affiliation(s)
- Anıl
Sera Çakmak
- Department
of Chemical Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Sümeyra Fuerkaiti
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Dilara Karagüzel
- Department
of Biology, Molecular Biology Section, Hacettepe
University, 06800 Ankara, Turkey
| | - Çağatay Karaaslan
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
- Department
of Biology, Molecular Biology Section, Hacettepe
University, 06800 Ankara, Turkey
| | - Menemşe Gümüşderelioğlu
- Department
of Chemical Engineering, Hacettepe University, 06800 Ankara, Turkey
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| |
Collapse
|
6
|
Fan J, Zhang X, Kang M, Lee CS, Kim L, Hadaya D, Aghaloo TL, Lee M. Complementary modulation of BMP signaling improves bone healing efficiency. Biomaterials 2023; 302:122335. [PMID: 37748419 PMCID: PMC10805245 DOI: 10.1016/j.biomaterials.2023.122335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/30/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
The bone morphogenetic protein (BMP) signaling pathway plays a crucial role in bone development and regeneration. While BMP-2 is widely used as an alternative to autograft, its clinical application has raised concerns about adverse side effects and deteriorated bone quality. Therefore, there is a need to develop more sophisticated approaches to regulate BMP signaling and promote bone regeneration. Here, we present a novel complementary strategy that targets both BMP antagonist noggin and agonist Trb3 to enhance bone defect repair without the application of exogenous BMP-2. In vitro studies showed that overexpression of Trb3 with simultaneous noggin suppression significantly promotes osteogenic differentiation of mesenchymal stem cells. This was accompanied by increased BMP/Smad signaling. We also developed sterosome nanocarriers, a non-phospholipid liposomal system, to achieve non-viral mediated noggin suppression and Trb3 overexpression. The gene-loaded sterosomes were integrated onto an apatite-coated polymer scaffold for in vivo calvarial defect implantation, resulting in robust bone healing compared to BMP-2 treatments. Our work provides a promising alternative for high-quality bone formation by regulating expression of BMP agonists and antagonists.
Collapse
Affiliation(s)
- Jiabing Fan
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, 21853, USA
| | - Xiao Zhang
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Minjee Kang
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Chung-Sung Lee
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA; Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Chungcheongnam-do, 31538, Republic of Korea
| | - Lauren Kim
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Danny Hadaya
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, USA
| | - Tara L Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, USA.
| | - Min Lee
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, 90095, USA; Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
7
|
Lee S, Chae DS, Song BW, Lim S, Kim SW, Kim IK, Hwang KC. ADSC-Based Cell Therapies for Musculoskeletal Disorders: A Review of Recent Clinical Trials. Int J Mol Sci 2021; 22:10586. [PMID: 34638927 PMCID: PMC8508846 DOI: 10.3390/ijms221910586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
Recently published clinical trials involving the use of adipose-derived stem cells (ADSCs) indicated that approximately one-third of the studies were conducted on musculoskeletal disorders (MSD). MSD refers to a wide range of degenerative conditions of joints, bones, and muscles, and these conditions are the most common causes of chronic disability worldwide, being a major burden to the society. Conventional treatment modalities for MSD are not sufficient to correct the underlying structural abnormalities. Hence, ADSC-based cell therapies are being tested as a form of alternative, yet more effective, therapies in the management of MSDs. Therefore, in this review, MSDs subjected to the ADSC-based therapy were further categorized as arthritis, craniomaxillofacial defects, tendon/ligament related disorders, and spine disorders, and their brief characterization as well as the corresponding conventional therapeutic approaches with possible mechanisms with which ADSCs produce regenerative effects in disease-specific microenvironments were discussed to provide an overview of under which circumstances and on what bases the ADSC-based cell therapy was implemented. Providing an overview of the current status of ADSC-based cell therapy on MSDs can help to develop better and optimized strategies of ADSC-based therapeutics for MSDs as well as help to find novel clinical applications of ADSCs in the near future.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 210-701, Korea;
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| |
Collapse
|
8
|
Zuo W, Yu L, Zhang H, Fei Q. Mineralized collagen scaffold bone graft accelerate the osteogenic process of HASCs in proper concentration. Regen Ther 2021; 18:161-167. [PMID: 34277898 PMCID: PMC8254075 DOI: 10.1016/j.reth.2021.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Purpose To investigate the feasibility and the optimum condition of human adipose-derived stem cells cultured on the mineralized collagen material; and to further explore the mechanism of osteogenic differentiation of the human Adipose-derived stem cells stimulated by the mineralized collagen material. Methods Primary human adipose-derived stem cells (HADSCs) were isolated from human adipose tissue using centrifugal stratification, which had been passed repeatedly to later generations and purified. Human adipose-derived stem cells were cultured on the bone graft material and the optimum concentration was explored by Alamar blue colorimetric method. The rest experiment was conducted according to the result. The experimental groups are shown below: group A (HADSCs + bone graft material); group B (HADSCs). Morphological observation was taken by scanning electronic microscope (SEM). Alkaline phosphatase activities were tested by histochemical method. Calcium deposition was investigated by alizarin red staining. The quantity access of osteogenic-related mRNA: ALP (alkaline phosphatase), BMP2 (bone morphogenetic protein 2) and RUNX2 (runt-related transcription factor 2) were detected using RT-PCR. Results The cultured cells grew stably and proliferated rapidly. The optimum condition was 0.5 mg/cm2 bone graft material coated on the bottom of medium. After culturing on the material 14 days, the alizarin red staining showed that more calcium deposition was detected in group A and alkaline phosphatase activities of group A was higher than group B (p ˃ 0.05). Similarly, after culturing for 14 days, the ALP, BMP2 and RUNX2 transcription activity of group A was higher than group B (p ˃ 0.05). Conclusion Human adipose-derived stem cells cultured on bone graft material were dominantly differentiated into osteoblast in vitro. Thus it provided a new choice for bone tissue engineering.
Collapse
Affiliation(s)
- Weiyang Zuo
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 100050, China
| | - Lingjia Yu
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 100050, China
| | - Haiyan Zhang
- Municipal Laboratory for Liver Protection and Regulation of Regeneration, Department of Cell Biology, Capital Medical University, Beijing, China
| | - Qi Fei
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 100050, China
| |
Collapse
|
9
|
Kim S, Fan J, Lee CS, Chen C, Lee M. Sulfonate Hydrogel-siRNA Conjugate Facilitates Osteogenic Differentiation of Mesenchymal Stem Cells by Controlled Gene Silencing and Activation of BMP Signaling. ACS APPLIED BIO MATERIALS 2021; 4:5189-5200. [PMID: 34661086 DOI: 10.1021/acsabm.1c00369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hydrogels have been widely used in bone tissue engineering due to their tunable characteristics that allow facile modifications with various biochemical properties to support cell growth and guide proper cell functions. Herein, we report a design of hydrogel-siRNA conjugate that facilitates osteogenesis via gene silencing and activation of bone morphogenetic protein (BMP) signaling. A sulfonate hydrogel is prepared by modifying chitosan with sulfoacetic acid to mimic a natural sulfated polysaccharide and to provide a hydrogel surface that enables BMP binding. Then, siRNA targeting noggin, an endogenous extracellular antagonist of BMP signaling, is covalently conjugated to the sulfonate hydrogel by visible blue light crosslinking. The sulfonate hydrogel-siRNA conjugate is efficient to bind BMPs and also successfully prolongs the release of siRNA for sustained noggin suppression, thereby resulting in significantly increased osteogenic differentiation. Lastly, demineralized bone matrix (DBM) is incorporated into the sulfonate hydrogel-siRNA conjugate, wherein the DBM incorporation induces noggin expression via a negative feedback mechanism that regulates BMP signaling in DBM. However, simultaneous delivery of siRNA downregulates noggin thus facilitating endogenous BMP activity and enhancing the osteogenic efficacy of DBM. These findings support a promising hydrogel RNA silencing platform for bone tissue engineering applications.
Collapse
Affiliation(s)
- Soyon Kim
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chung-Sung Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Chen Chen
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA.,Department of Bioengineering, University of California, Los Angeles, USA
| |
Collapse
|
10
|
Choi LY, Kim MH, Nam YK, Kim JH, Cho HY, Yang WM. Palmul-Tang, a Korean Medicine, Promotes Bone Formation via BMP-2 Pathway in Osteoporosis. Front Pharmacol 2021; 12:643482. [PMID: 33841161 PMCID: PMC8032944 DOI: 10.3389/fphar.2021.643482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/19/2021] [Indexed: 12/30/2022] Open
Abstract
Osteoporosis is a common skeletal disease in post-menopausal women. Palmul-tang, an herbal medicine, has been treated for gynecological disease such as anemia, anorexia, anti-fatigue, unspecified menstruation and female infertility in East Asia. In this study, ameliorative effects of Palmul-tang soft extracts (PMT), a Korean Medicine, on osteoporosis were investigated. Ovariectomized (OVX) osteoporotic ICR mice were intragastrically administrated PMT for 4 weeks. The level of bone mineral density (BMD) was analyzed in bone tissues by dual X-ray absorptiometry. The bone medullary cavity and deposition of collagen were investigated by histological analysis. In addition, the BMP-2 signaling-related molecules, osteoblastic differentiation and formation markers, were determined in femoral tissues. The levels of BMD and bone mineral content were significantly increased in tibia, femurs and LV by treatment of PMT. PMT replenished bone marrow cavity and increased collagen deposition in bone marrow cells of femur. In addition, administration of PMT recovered serum ALP, bALP, osteocalcin and calcium levels in osteoporotic mice. Moreover, PMT treatment up-regulated the expressions of BMP-2, RUNX2 and OSX with its downstream factors, ALP, OPN and BSP-1, in the femoral tissues. Taken together, PMT restored the bone minerals and improvement of bone integrity by bone-forming BMP-2 signaling pathway. These results demonstrate that PMT could be an ameliorative agent for osteoporosis.
Collapse
Affiliation(s)
- La Yoon Choi
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Mi Hye Kim
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Yeon Kyung Nam
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Ju Hee Kim
- College of Pharmacy, CHA University, Seongnam, South Korea
| | - Hea-Young Cho
- College of Pharmacy, CHA University, Seongnam, South Korea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
11
|
Wu Z, Wang Y, Han W, Yang K, Hai E, Ma R, Di Z, Shang F, Su R, Wang R, Wang Z, Zhang Y, Li J. EDA and EDAR expression at different stages of hair follicle development in cashmere goats and effects on expression of related genes. Arch Anim Breed 2020; 63:461-470. [PMID: 33473371 PMCID: PMC7810227 DOI: 10.5194/aab-63-461-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/16/2020] [Indexed: 11/25/2022] Open
Abstract
This study is focused on the detection of ectodysplasin A (EDA) and ectodysplasin A receptor
(EDAR) mRNA expression levels and protein positions in seven stages of
cashmere goat fetus development (45, 55, 65, 75 95, 115, and 135 d), with the main goal of
investigating the effect of EDA and EDAR on genes related to hair follicle
development.
Quantitative real-time polymerase chain reaction (RT-qPCR) was used to
measure EDA and EDAR expression levels in seven stages of cashmere goat
fetus development. Immunohistochemistry (IHC) was used to locate EDA and EDAR
in the critical stage of fetal hair follicle development (45–135 d). EDA and EDAR expression in fetal fibroblasts and epithelial cells was
interfered with by short hairpin RNA (sh-RNA). The results indicated that
EDA and EDAR were both expressed in the skin tissue in the seven cashmere
goat embryo stages. Moreover, EDA and EDAR play an important role in the
formation of embryonic placode (Pc). After interfering with EDA and EDAR,
the expression of BMP2, BMP4, noggin, β-catenin, TGF-β2,
Wnt-10b, and NOTCH1 in fibroblasts and epithelial cells changed
significantly.
This study provides a theoretical and
experimental basis for further studying the molecular regulation mechanism
of hair follicle development.
Collapse
Affiliation(s)
- Zhihong Wu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Yu Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Wenjing Han
- College of Chemistry and Life Science, Chifeng University, Chifeng, Inner Mongolia Autonomous Region 024000, China
| | - Kun Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Erhan Hai
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Rong Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Zhengyang Di
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Fangzheng Shang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Ruijun Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region 010018, China
| | - Jinquan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010018, China.,Engineering Research Center for Goat Genetics and Breeding, Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region 010018, China.,Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot, Inner Mongolia Autonomous Region 010018, China
| |
Collapse
|
12
|
Dong S, Li J, Zhang X. Tumor protein p53-induced nuclear protein 2 modulates osteogenic differentiation of human adipose derived stem/stromal cells by activating Wnt/β-catenin signaling. Am J Transl Res 2020; 12:6853-6867. [PMID: 33194077 PMCID: PMC7653607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/25/2020] [Indexed: 06/11/2023]
Abstract
Human adipose derived stem/stromal cells (hASCs) are frequently used as seed cells in bone tissue engineering. These cells have good osteogenic properties in various in vivo and in vitro models. Tumor protein p53-induced nuclear protein 2 (TP53INP2) regulates apoptosis, autophagy, and cell differentiation. However, whether TP53INP2 regulates osteogenic differentiation of hASCs has not been sufficiently studied. Herein, we explored this topic using siRNA experiments, osteogenic induction, quantitative real-time PCR (qRT-PCR) and western blot analysis. We found that siRNA decreased mRNA levels of osteoblast-specific genes in TP53INP2 cells. Western blots showed that RUNX2 protein expression decreased in siRNA-TP53INP2 cells at day 3, 7, and 21 after osteogenic induction. The level of β-catenin, LC3 and the LC3-II/LC3-I ratio in siRNA-TP53INP2 cells was decreased at day 3 and 7 after osteogenic induction. Further, treatment with lithium chloride (LiCl), an activator of Wnt signaling pathway, induced partial recovery of protein expression of β-catenin and RUNX2 (osteoblast-specific factor 2) in TP53INP2 knockdown cells. Collectively, these results show that TP53INP2 promotes osteogenic differentiation of hASCs by activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Shi Dong
- College of Stomatology, Chongqing Medical UniversityChongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical UniversityChongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing, China
| | - Xiaonan Zhang
- College of Stomatology, Chongqing Medical UniversityChongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing, China
| |
Collapse
|
13
|
Bez M, Pelled G, Gazit D. BMP gene delivery for skeletal tissue regeneration. Bone 2020; 137:115449. [PMID: 32447073 PMCID: PMC7354211 DOI: 10.1016/j.bone.2020.115449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
Musculoskeletal disorders are common and can be associated with significant morbidity and reduced quality of life. Current treatments for major bone loss or cartilage defects are insufficient. Bone morphogenetic proteins (BMPs) are key players in the recruitment and regeneration of damaged musculoskeletal tissues, and attempts have been made to introduce the protein to fracture sites with limited success. In the last 20 years we have seen a substantial progress in the development of various BMP gene delivery platforms for several conditions. In this review we cover the progress made using several techniques for BMP gene delivery for bone as well as cartilage regeneration, with focus on recent advances in the field of skeletal tissue engineering. Some methods have shown success in large animal models, and with the global trend of introducing gene therapies into the clinical setting, it seems that the day in which BMP gene therapy will be viable for clinical use is near.
Collapse
Affiliation(s)
- Maxim Bez
- Medical Corps, Israel Defense Forces, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Gadi Pelled
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA; Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Dan Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA; Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
14
|
May RD, Frauchiger DA, Albers CE, Tekari A, Benneker LM, Klenke FM, Hofstetter W, Gantenbein B. Application of Cytokines of the Bone Morphogenetic Protein (BMP) Family in Spinal Fusion - Effects on the Bone, Intervertebral Disc and Mesenchymal Stromal Cells. Curr Stem Cell Res Ther 2020; 14:618-643. [PMID: 31455201 PMCID: PMC7040507 DOI: 10.2174/1574888x14666190628103528] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is a prevalent socio-economic burden and is often associated with damaged or degenerated intervertebral discs (IVDs). When conservative therapy fails, removal of the IVD (discectomy), followed by intersomatic spinal fusion, is currently the standard practice in clinics. The remaining space is filled with an intersomatic device (cage) and with bone substitutes to achieve disc height compensation and bone fusion. As a complication, in up to 30% of cases, spinal non-fusions result in a painful pseudoarthrosis. Bone morphogenetic proteins (BMPs) have been clinically applied with varied outcomes. Several members of the BMP family, such as BMP2, BMP4, BMP6, BMP7, and BMP9, are known to induce osteogenesis. Questions remain on why hyper-physiological doses of BMPs do not show beneficial effects in certain patients. In this respect, BMP antagonists secreted by mesenchymal cells, which might interfere with or block the action of BMPs, have drawn research attention as possible targets for the enhancement of spinal fusion or the prevention of non-unions. Examples of these antagonists are noggin, gremlin1 and 2, chordin, follistatin, BMP3, and twisted gastrulation. In this review, we discuss current evidence of the osteogenic effects of several members of the BMP family on osteoblasts, IVD cells, and mesenchymal stromal cells. We consider in vitro and in vivo studies performed in human, mouse, rat, and rabbit related to BMP and BMP antagonists in the last two decades. We give insights into the effects that BMP have on the ossification of the spine. Furthermore, the benefits, pitfalls, and possible safety concerns using these cytokines for the improvement of spinal fusion are discussed.
Collapse
Affiliation(s)
- Rahel Deborah May
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Christoph Emmanuel Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Lorin Michael Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Frank Michael Klenke
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
15
|
Lee CS, Kim S, Fan J, Hwang HS, Aghaloo T, Lee M. Smoothened agonist sterosome immobilized hybrid scaffold for bone regeneration. SCIENCE ADVANCES 2020; 6:eaaz7822. [PMID: 32494652 PMCID: PMC7176430 DOI: 10.1126/sciadv.aaz7822] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/28/2020] [Indexed: 05/24/2023]
Abstract
Biomaterial delivery of bioactive agents and manipulation of stem cell fate are an attractive approach to promote tissue regeneration. Here, smoothened agonist sterosome is developed using small-molecule activators [20S-hydroxycholesterol (OHC) and purmorphamine (PUR)] of the smoothened protein in the hedgehog pathway as carrier and cargo. Sterosome presents inherent osteoinductive property even without drug loading. Sterosome is covalently immobilized onto three-dimensional scaffolds via a bioinspired polydopamine intermediate to fabricate a hybrid scaffold for bone regeneration. Sterosome-immobilized hybrid scaffold not only provides a favorable substrate for cell adhesion and proliferation but also delivers bioactive agents in a sustained and spatially targeted manner. Furthermore, this scaffold significantly improves osteogenic differentiation of bone marrow stem cells through OHC/PUR-mediated synergistic activation of the hedgehog pathway and also enhances bone repair in a mouse calvarial defect model. This system serves as a versatile biomaterial platform for many applications, including therapeutic delivery and endogenous regenerative medicine.
Collapse
Affiliation(s)
- Chung-Sung Lee
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Soyon Kim
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Hee Sook Hwang
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California at Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Alfotawi R, Elsafadi M, Muthurangan M, Siyal AA, Alfayez M, Mahmmod AA. A New Procedure in Bone Engineering Using Induced Adipose Tissue. J INVEST SURG 2019; 34:44-54. [PMID: 31558065 DOI: 10.1080/08941939.2019.1604915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background: Osteoporosis is associated with a metabolic imbalance between adipogenesis and osteogenesis. We hypothesized that implanting a carrier for differentiated stem cells and signaling molecules inside adipose tissues could be used to enable transdifferentiation between cells, upregulate osteogenesis, and support bone formation, which may regain the balance between osteogenesis and adipogenesis. Methodology: A CL1 human mesenchymal stem cell line was grown in an osteogenic medium to differentiate into osteoblasts, and the differentiated cells were then exposed to an adipogenic medium to stimulate differentiation into adipocytes. Osteogenic and adipogenic differentiation were confirmed by the following assays: alkaline phosphatase staining, Nile red Staining, and quantitative real-time polymerase chain reaction (qPCR). The ratio of adipocytes to osteocytes for both cases was calculated. To evaluate bone induction in vivo, a calcium sulfate/hydroxyapatite cement was prepared in a syringe and then seeded with 106 cells/mL of rat bone marrow stromal cells (rMSCs) and covered with 1 mL of tissue culture media containing 0.1 mg of bone morphogenetic protein 7 (BMP-7). The construct was injected into the abdominal fat tissue of 10 male Sprague-Dawley rats. Results: The conversion of osteocytes to adipocytes was 20-fold greater than the reverse conversion, and the area of bone regeneration was 15.7 ± 3.7%, the area of adipose tissue was 65.8 ± 13.1%, and the area of fibrous tissue was 18.3 ± 7.8%. Conclusion: Adipogenic interconversion and associated bone formation demonstrate the potential of a new therapy for balancing osteogenesis and adipogenesis.
Collapse
Affiliation(s)
- Randa Alfotawi
- Oral and Maxillofacial Department, King Saud University, Riyadh, Saudi Arabia
| | - Mona Elsafadi
- Stem Cell Unit, Anatomy Department, Medical School, King Saud University, Riyadh, Saudi Arabia
| | - Manikandan Muthurangan
- Stem Cell Unit, Anatomy Department, Medical School, King Saud University, Riyadh, Saudi Arabia
| | - Abdul-Aziz Siyal
- Stem Cell Unit, Anatomy Department, Medical School, King Saud University, Riyadh, Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Anatomy Department, Medical School, King Saud University, Riyadh, Saudi Arabia
| | - Amer A Mahmmod
- Stem Cell Unit, Anatomy Department, Medical School, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Review of the Pathways Involved in the Osteogenic Differentiation of Adipose-Derived Stem Cells. J Craniofac Surg 2019; 30:703-708. [PMID: 30839467 DOI: 10.1097/scs.0000000000005447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Grafts and prosthetic materials used for the repair of bone defects are often accompanied by comorbidity and rejection. Therefore, there is an immense need for novel approaches to combating the issues surrounding such defects. Because of their accessibility, substantial proportion, and osteogenic differentiation potential, adipose-derived stem cells (ASCs) make for an ideal source of bone tissue in regenerative medicine. However, efficient induction of ASCs toward an osteoblastic lineage in vivo is met with challenges, and many signaling pathways must come together to secure osteoblastogenesis. Among them are bone morphogenic protein, wingless-related integration site protein, Notch, Hedgehog, fibroblast growth factor, vascular endothelial growth factor, and extracellular regulated-signal kinase. The goal of this literature review is to conglomerate the present research on these pathways to formulate a better understanding of how ASCs are most effectively transformed into bone in the context of tissue engineering.
Collapse
|
18
|
Huang M, Zhang X, Li J, Li Y, Wang Q, Teng W. Comparison of osteogenic differentiation induced by siNoggin and pBMP-2 delivered by lipopolysaccharide-amine nanopolymersomes and underlying molecular mechanisms. Int J Nanomedicine 2019; 14:4229-4245. [PMID: 31239677 PMCID: PMC6559258 DOI: 10.2147/ijn.s203540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose: Gene therapies via Noggin small interfering (si)RNA (siNoggin) and bone morphogenetic protein (BMP)-2 plasmid DNA (pBMP-2) may be promising strategies for bone repair/regeneration, but their ideal delivery vectors, efficacy difference, and underlying mechanisms have not been explored, so these issues were probed here. Methods: This study used lipopolysaccharide-amine nanopolymersomes (LNPs), an efficient cytosolic delivery vector developed by the research team, to mediate siNoggin and pBMP-2 to transfect MC3T3-E1 cells, respectively. The cytotoxicity, cell uptake, and gene knockdown efficiency of siNoggin-loaded LNPs (LNPs/siNoggin) were studied, then the osteogenic-differentiation efficacy of MC3T3-E1 cells treated by LNPs/pBMP-2 and LNPs/siNoggin, respectively, were compared by measuring the expression of osteogenesis-related genes and proteins, alkaline phosphatase (ALP) activity, and mineralization of the extracellular matrix at all osteogenic stages. Finally, the possible signaling pathways of the two treatments were explored. Results: LNPs delivered siNoggin into cells efficiently to silence 50% of Noggin expression without obvious cytotoxicity. LNPs/siNoggin and LNPs/pBMP-2 enhanced the osteogenic differentiation of MC3T3 E1 cells, but LNPs/siNoggin was better than LNPs/pBMP-2. BMP/Mothers against decapentaplegic homolog (Smad) and glycogen synthase kinase (GSK)-3β/β-catenin signaling pathways appeared to be involved in osteogenic differentiation induced by LNPs/siNoggin, but GSK-3β/β-catenin was not stimulated upon LNPs/pBMP-2 treatment. Conclusion: LNPs are safe and efficient delivery vectors for DNA and RNA, which may find wide applications in gene therapy. siNoggin treatment may be a more efficient strategy to enhance osteogenic differentiation than pBMP-2 treatment. LNPs loaded with siNoggin and/or pBMP-2 may provide new opportunities for the repair and regeneration of bone.
Collapse
Affiliation(s)
- Mingdi Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinchun Zhang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Li
- Laboratory of Biomaterials, Key Laboratory on Assisted Circulation, Ministry of Health, Cardiovascular Division, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanshan Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qinmei Wang
- Laboratory of Biomaterials, Key Laboratory on Assisted Circulation, Ministry of Health, Cardiovascular Division, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Teng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
19
|
Damanik FFR, Spadolini G, Rotmans J, Farè S, Moroni L. Biological activity of human mesenchymal stromal cells on polymeric electrospun scaffolds. Biomater Sci 2019; 7:1088-1100. [PMID: 30633255 DOI: 10.1039/c8bm00693h] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Electrospinning provides a simple robust method to manufacture scaffolds for tissue engineering applications. Though varieties of materials can be used, optimization and biocompatibility tests are required to provide functional tissue regeneration. Moreover, many studies are limited to 2D electrospun constructs rather than 3D templates due to the production of high density packed fibres, which result in poor cell infiltration. Here, we optimised electrospinning parameters for three different polymers: poly(ε-caprolactone) (PCL), polylactic acid (PLA) and poly(ethylene oxide terephthalate)/poly(butylene terephthalate) (PA) copolymers. Human mesenchymal stromal cells (hMSCs) were cultured on scaffolds for 14 days to study the scaffolds' biocompatibility and their multi-lineage differentiation potential or maintenance of stemness in the absence of chemical stimuli. For all scaffolds, a high and stable metabolic activity was measured throughout the culture time with a high proliferation rate compared to day 1 (PCL 5.8-, PLA 4-, PA 4.9-fold). The metabolism of hMSCs was also measured through glucose and lactate concentrations, showing no cytotoxic levels up to 14 days. Total glycosaminoglycan (GAG) production was the highest in PA electrospun scaffolds. When normalized to DNA, GAG production was the highest in PLA and PA scaffolds. All scaffolds were prone to differentiate to an osteogenic lineage, with PCL providing the highest alkaline phosphatase and collagen type Ia gene upregulation. As PA had the most stable fibre formation, it was chosen as a template to further incorporate stromal cell-derived factor-1 (SDF-1) and granulocyte colony-stimulating factor (G-CSF), and stimulate higher hMSC infiltration. These scaffolds provided significantly higher hMSC infiltration than normal PA scaffolds. In conclusion, our optimized biocompatible electrospun scaffolds have shown promising regulation of hMSC fate. When combined with migratory stimulating cytokines, these scaffolds may overcome the known challenges of poor cellular infiltration typical of micro- and nano-fibrillary random meshes.
Collapse
Affiliation(s)
- Febriyani F R Damanik
- University of Twente, Drienerlolaan 5, Zuidhorst 145, 7522 NB Enschede, the Netherlands
| | | | | | | | | |
Collapse
|
20
|
Hui T, Zhang GC, Feng DD, Ji P. [Role of neuropeptide substance P and the bone morphogenetic protein signaling pathway in osteogenic differentiation of ST2 cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 36:378-383. [PMID: 30182564 DOI: 10.7518/hxkq.2018.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study aimed to investigate the role and mechanism of neuropeptide substance P (SP) in ST2 cell (bone mesenchymal stem cells of mice) osteogenic differentiation to provide a basis for the treatment of temporomandibular joint osteoarthritis. METHODS Third-generation ST2 cells were cultured with different concentrations of SP (0, 10⁻¹⁰, 10⁻⁸, 10⁻⁶, and 10⁻⁵ mol·L⁻¹). After 24, 48, and 72 h, cell proliferation was detected by CCK-8. The ST2 cells were cultured with 10⁻⁶ mol·L⁻¹ SP for 1, 3, 5, and 7 days. Subsequently, the expression of alkaline phosphatase (ALP), collagen typeⅠ(CollaⅠ), and osteocalcin (OCN) in the culture supernatant was tested by enzyme-linked immunosorbent assay (ELISA). ALP activity was detected by immunofluorescence staining. The ST2 cells were cultured with SP, Noggin (inhibitor of the bone morphogenetic protein signaling pathway), SP+Noggin, and 2% fetal bovine serum, respectively. Finally, the expression of ALP, CollaⅠ, and OCN in the culture supernatant was tested by ELISA. RESULTS CCK-8 showed that the effect of cell proliferation was most obvious when the SP concentration was 10⁻⁶ mol·L⁻¹ (P<0.01). The ELISA results demonstrated that ALP expression significantly increased at day 5 compared with that in the control group (P<0.01), whereas the expression of CollaⅠand OCN significantly increased at day 7 (P<0.05). Immunofluorescence results showed that ALP activity was strongest at day 5. The expression of ALP, CollaⅠ, and OCN decreased after Noggin addition (P<0.05). CONCLUSIONS SP can promote the proliferation and osteogenic differentiation of ST2 cells, and the bone morphogenetic protein signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Ting Hui
- Dept. of Prosthodontics, Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan 250012, China
| | - Guang-Can Zhang
- Dept. of Prosthodontics, Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan 250012, China
| | - Dan-Dan Feng
- Dept. of Prosthodontics, Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan 250012, China
| | - Ping Ji
- Dept. of Prosthodontics, Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan 250012, China
| |
Collapse
|
21
|
Convergence of TGFβ and BMP signaling in regulating human bone marrow stromal cell differentiation. Sci Rep 2019; 9:4977. [PMID: 30899078 PMCID: PMC6428815 DOI: 10.1038/s41598-019-41543-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/26/2019] [Indexed: 01/11/2023] Open
Abstract
Targeting regulatory signaling pathways that control human bone marrow stromal (skeletal or mesenchymal) stem cell (hBMSC) differentiation and lineage fate determination is gaining momentum in the regenerative medicine field. Therefore, to identify the central regulatory mechanism of osteoblast differentiation of hBMSCs, the molecular phenotypes of two clonal hBMSC lines exhibiting opposite in vivo phenotypes, namely, bone forming (hBMSC+bone) and non-bone forming (hBMSC−Bone) cells, were studied. Global transcriptome analysis revealed significant downregulation of several TGFβ responsive genes, namely, TAGLN, TMP1, ACTA2, TGFβ2, SMAD6, SMAD9, BMP2, and BMP4 in hBMSC−Bone cells and upregulation on SERPINB2 and NOG. Transcriptomic data was associated with marked reduction in SMAD2 protein phosphorylation, which thereby implies the inactivation of TGFβ and BMP signaling in those cells. Concordantly, activation of TGFβ signaling in hBMSC−Bone cells using either recombinant TGFβ1 protein or knockdown of SERPINB2 TGFβ-responsive gene partially restored their osteoblastic differentiation potential. Similarly, the activation of BMP signaling using exogenous BMP4 or via siRNA-mediated knockdown of NOG partially restored the differentiation phenotype of hBMSC−Bone cells. Concordantly, recombinant NOG impaired ex vivo osteoblastic differentiation of hBMSC+Bone cells, which was associated with SERBINB2 upregulation. Our data suggests the existence of reciprocal relationship between TGFB and BMP signaling that regulates hBMSC lineage commitment and differentiation, whilst provide a plausible strategy for generating osteoblastic committed cells from hBMSCs for clinical applications.
Collapse
|
22
|
Kim S, Cui ZK, Koo B, Zheng J, Aghaloo T, Lee M. Chitosan-Lysozyme Conjugates for Enzyme-Triggered Hydrogel Degradation in Tissue Engineering Applications. ACS APPLIED MATERIALS & INTERFACES 2018; 10:41138-41145. [PMID: 30421603 PMCID: PMC6453716 DOI: 10.1021/acsami.8b15591] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Tuning hydrogel degradation enables effective and successful tissue regeneration by modulating cellular behaviors and matrix formation. In this work, we develop a novel degradable hydrogel scaffold on the basis of a unique enzyme-substrate complex by photocrosslinking. Chitosan and lysozyme are chemically modified with methacrylate moieties to be tethered in hydrogels, and in the presence of riboflavin initiator, these hydrogels are cured by blue light irradiation. The incorporation of lysozyme to chitosan hydrogels accelerates the degradation rate of the crosslinked hydrogels in a dose-dependent manner, as evidenced by an increase in pore size and interconnectivity through cryogenic scanning electron microscopy over time. Those noncytotoxic materials significantly enhance cellular proliferation and migration, which contribute to osteogenic differentiation of encapsulated mesenchymal stem cells in vitro and bone formation in mouse calvarial defects. These findings suggest a promising strategy to modulate the degradation behavior of hydrogels for use in tissue engineering.
Collapse
Affiliation(s)
- Soyon Kim
- Department of Bioengineering, University of California, Los Angeles, USA
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
| | - Bonhye Koo
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, USA
| | - Jiwen Zheng
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, USA
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, University of California, Los Angeles, USA
| | - Min Lee
- Department of Bioengineering, University of California, Los Angeles, USA
- Division of Advanced Prosthodontics, University of California, Los Angeles, USA
- Corresponding author: Min Lee, PhD, UCLA School of Dentistry, 10833 Le Conte Avenue, CHS 23-088F, Los Angeles, CA 90095-1668, USA, , Phone: +1-310-825-6674, Fax: +1-310-825-6345
| |
Collapse
|
23
|
Ebnerasuly F, Hajebrahimi Z, Tabaie SM, Darbouy M. Simulated Microgravity Condition Alters the Gene Expression of some ECM and Adhesion Molecules in Adipose Derived Stem Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:146-157. [PMID: 31565646 PMCID: PMC6744620 DOI: 10.22088/ijmcm.bums.7.3.146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/27/2018] [Indexed: 01/05/2023]
Abstract
Adipose- derived stem cells (ADSCs) are widely used for tissue engineering and regenerative medicine. The beneficial effects of ADSCs on wound healing have already been reported. Remodeling of extracellular matrix (ECM) is the most important physiological event during wound healing. ECM is sensitive to mechanical stresses and the expression of its components can be therefore influenced. The aim of this study was to investigate the effect of simulated microgravity on gene expression of some ECM and adhesion molecules in human ADSCs. After isolation and characterization of ADSCs, cells were exposed to simulated microgravity for 1, 3 and 7 days. Real-time PCR, fluorescence immunocytochemistry, and MTT assay were performed to evaluate the alterations of integrin subunit beta 1 (ITGB1), collagen type 3 (ColIII), matrix metalloproteinase-1 (MMP1), CD44, fibrillin (FBN1), vimentin (VIM) genes, and ColIII protein levels as well as cells viability. Microgravity simulation increased the expression of ITGB1, ColIII, MMP1, and CD44 and declined the expression of FBN1 and VIM genes. ColIII protein levels also increased. There were no significant changes in the viability of cells cultured in microgravity. Since the high expression of ECM components is known as one of the fibroblast markers, our data suggest that pretreatment of ADSCs by simulated microgravity may increase their differentiation capacity towards fibroblastic cells. Microgravity had not adversely affected the viability of ADSCs, and it is likely to be used alone or in combination with biochemical inducers for cell manipulation.
Collapse
Affiliation(s)
- Farid Ebnerasuly
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Zahra Hajebrahimi
- Aerospace Research Institute, Ministry of Science Research and Technology, Tehran, Iran
| | - Seyed Mehdi Tabaie
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Mojtaba Darbouy
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran.,Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
24
|
Low-intensity pulsed ultrasound stimulation facilitates in vitro osteogenic differentiation of human adipose-derived stem cells via up-regulation of heat shock protein (HSP)70, HSP90, and bone morphogenetic protein (BMP) signaling pathway. Biosci Rep 2018; 38:BSR20180087. [PMID: 29789443 PMCID: PMC6048203 DOI: 10.1042/bsr20180087] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 02/01/2023] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has positive effects on osteogenic differentiation. However, the effect of LIPUS on osteogenic differentiation of human adipose-derived stem cells (hASCs) is unclear. In the present study, we investigated whether LIPUS could promote the proliferation and osteogenic differentiation of hASCs. hASCs were isolated and osteogenically induced with LIPUS stimulation at 20 and 30 mW cm-2 for 30 min day-1 Cell proliferation and osteogenic differentiation potential of hASCs were respectively analyzed by cell counting kit-8 assay, Alizarin Red S staining, real-time polymerase chain reaction, and Western blotting. The results indicated that LIPUS stimulation did not significantly affect the proliferation of hASCs, but significantly increased their alkaline phosphatase activity on day 6 of culture and markedly promoted the formation of mineralized nodules on day 21 of culture. The mRNA expression levels of runt-related transcription factor, osteopontin, and osteocalcin were significantly up-regulated by LIPUS stimulation. LIPUS stimulation did not affect the expression of heat shock protein (HSP) 27, HSP40, bone morphogenetic protein (BMP)-6 and BMP-9, but significantly up-regulated the protein levels of HSP70, HSP90, BMP-2, and BMP-7 in the hASCs. Further studies found that LIPUS increased the mRNA levels of Smad 1 and Smad 5, elevated the phosphorylation of Smad 1/5, and suppressed the expression of BMP antagonist Noggin. These findings indicated that LIPUS stimulation enhanced osteogenic differentiation of hASCs possibly through the up-regulation of HSP70 and HSP90 expression and activation of BMP signaling pathway. Therefore, LIPUS might have the potential to promote the repair of bone defect.
Collapse
|
25
|
Inhibitory Effects of Human Primary Intervertebral Disc Cells on Human Primary Osteoblasts in a Co-Culture System. Int J Mol Sci 2018; 19:ijms19041195. [PMID: 29652862 PMCID: PMC5979604 DOI: 10.3390/ijms19041195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 11/16/2022] Open
Abstract
Spinal fusion is a common surgical procedure to address a range of spinal pathologies, like damaged or degenerated discs. After the removal of the intervertebral disc (IVD), a structural spacer is positioned followed by internal fixation, and fusion of the degenerated segment by natural bone growth. Due to their osteoinductive properties, bone morphogenetic proteins (BMP) are applied to promote spinal fusion. Although spinal fusion is successful in most patients, the rates of non-unions after lumbar spine fusion range from 5% to 35%. Clinical observations and recent studies indicate, that the incomplete removal of disc tissue might lead to failure of spinal fusion. Yet, it is still unknown if a secretion of BMP antagonists in intervertebral disc (IVD) cells could be the reason of inhibition in bone formation. In this study, we co-cultured human primary osteoblasts (OB) and IVD cells i.e., nucleus pulposus (NPC), annulus fibrosus (AFC) and cartilaginous endplate cells (CEPC), to test the possible inhibitory effect from IVD cells on OB. Although we could see a trend in lower matrix mineralization in OB co-cultured with IVD cells, results of alkaline phosphatase (ALP) activity and gene expression of major bone genes were inconclusive. However, in NPC, AFC and CEPC beads, an up-regulation of several BMP antagonist genes could be detected. Despite being able to show several indicators for an inhibition of osteoinductive effects due to IVD cells, the reasons for pseudarthrosis after spinal fusion remain unclear.
Collapse
|
26
|
Li KC, Chang YH, Hsu MN, Lo SC, Li WH, Hu YC. Baculovirus-Mediated miR-214 Knockdown Shifts Osteoporotic ASCs Differentiation and Improves Osteoporotic Bone Defects Repair. Sci Rep 2017; 7:16225. [PMID: 29176755 PMCID: PMC5701180 DOI: 10.1038/s41598-017-16547-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Osteoporotic patients often suffer from bone fracture but its healing is compromised due to impaired osteogenesis potential of bone marrow-derived mesenchymal stem cells (BMSCs). Here we aimed to exploit adipose-derived stem cells from ovariectomized rats (OVX-ASCs) for bone healing. We unraveled that OVX-ASCs highly expressed miR-214 and identified 2 miR-214 targets: CTNNB1 (β-catenin) and TAB2. We demonstrated that miR-214 targeting of these two genes blocked the Wnt pathway, led to preferable adipogenesis and hindered osteogenesis. As a result, OVX-ASCs implantation into OVX rats failed to heal critical-size metaphyseal bone defects. We further engineered the OVX-ASCs with a novel Cre/loxP-based hybrid baculovirus vector that conferred prolonged expression of miR-214 sponge. Gene delivery for miR-214 sponge expression successfully downregulated miR-214 levels, activated the Wnt pathway, upregulated osteogenic factors β-catenin/Runx2, downregulated adipogenic factors PPAR-γ and C/EBP-α, shifted the differentiation propensity towards osteogenic lineage, enhanced the osteogenesis of co-cultured OVX-BMSCs, elevated BMP7/osteoprotegerin secretion and hindered exosomal miR-214/osteopontin release. Consequently, implanting the miR-214 sponge-expressing OVX-ASCs tremendously improved bone healing in OVX rats. Co-expression of miR-214 sponge and BMP2 further synergized the OVX-ASCs-mediated bone regeneration in OVX rats. This study implicates the potential of suppressing miR-214 by baculovirus-mediated gene delivery in osteoporotic ASCs for regenerative medicine.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Yu-Han Chang
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
- Department of Orthopaedic, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Shih-Chun Lo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Wan-Hua Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan.
| |
Collapse
|
27
|
Sun XK, Zhou J, Zhang L, Ma T, Wang YH, Yang YM, Tang YT, Li H, Wang LJ. Down-regulation of Noggin and miR-138 coordinately promote osteogenesis of mesenchymal stem cells. J Mol Histol 2017; 48:427-436. [PMID: 29094227 DOI: 10.1007/s10735-017-9740-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/22/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate to osteocytes under suitable conditions. In recent years, micro-nucleotides have been progressively used to modulate gene expression in cells due to the consideration of safety. Our present study aimed to investigate whether co-delivery of Noggin-siRNA and antimiR-138 enhances the osteogenic effect of MSCs. Using a murine MSC line, C3H/10T1/2 cells, the delivery efficiency of Noggin-siRNA and antimiR-138 into MSCs was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). Cell phenotype and proliferation capacity was assessed by flow cytometry and MTT assay respectively. The osteogenesis of MSCs was tested by Alkaline Phosphatase (ALP) staining, qRT-PCR, and western blot analyses. Our results demonstrated that the expression of Noggin and miR-138 were significantly silenced in MSCs by Noggin-siRNA and/or antimiR-138 delivery, while the phenotype and proliferation capacity of MSCs were not affected. Down-regulation of Noggin and miR-138 cooperatively promoted osteogenic differentiation of MSCs. The ALP positive cells reached about 83.57 ± 10.18%. Compared with single delivery, the expression of osteogenic related genes, such as Alp, Col-1, Bmp2, Ocn and Runx2, were the highest in cells with co-delivery of the two oligonucleotides. Moreover, the protein level of RUNX2, and the ratios of pSMAD1/5/SMAD1/5 and pERK1/2/ERK1/2 were significantly increased. The activation of Smad, Erk signaling may constitute the underlying mechanism of the enhanced osteogenesis process. Taken together, our study provides a safe strategy for the clinical rehabilitation application of MSCs in skeletal deficiency.
Collapse
Affiliation(s)
- Xing-Kun Sun
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China
- Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China
| | - Lei Zhang
- School of Biological and Chemical Engineering, ZheJiang University of Science & Technology, Hangzhou, 310023, China
| | - Tian Ma
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu-Han Wang
- Tibet Vocational Technical College, Lhasa, 850032, Tibet Autonomous Region, China
| | - Yan-Mei Yang
- Department of Stomatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan-Ting Tang
- Department of Stomatology, People's Hospital of Suzhou High-tech Zone, Suzhou, 215129, Jiangsu Province, China
| | - Hong Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China.
| | - Li-Jun Wang
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China.
| |
Collapse
|
28
|
Hypoxia Is a Critical Parameter for Chondrogenic Differentiation of Human Umbilical Cord Blood Mesenchymal Stem Cells in Type I/III Collagen Sponges. Int J Mol Sci 2017; 18:ijms18091933. [PMID: 28885597 PMCID: PMC5618582 DOI: 10.3390/ijms18091933] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/29/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022] Open
Abstract
Umbilical cord blood (UCB) is an attractive alternative to bone marrow for isolation of mesenchymal stem cells (MSCs) to treat articular cartilage defects. Here, we set out to determine the growth factors (bone morphogenetic protein 2 (BMP-2) and transforming growth factor-β (TGF-β1)) and oxygen tension effects during chondrogenesis of human UCB-MSCs for cartilage engineering. Chondrogenic differentiation was induced using 3D cultures in type I/III collagen sponges with chondrogenic factors in normoxia (21% O₂) or hypoxia (<5% O₂) for 7, 14 and 21 days. Our results show that UCB-MSCs can be committed to chondrogenesis in the presence of BMP-2+TGF-β1. Normoxia induced the highest levels of chondrocyte-specific markers. However, hypoxia exerted more benefit by decreasing collagen X and matrix metalloproteinase-13 (MMP13) expression, two chondrocyte hypertrophy markers. However, a better chondrogenesis was obtained by switching oxygen conditions, with seven days in normoxia followed by 14 days in hypoxia, since these conditions avoid hypertrophy of hUCB-MSC-derived chondrocytes while maintaining the expression of chondrocyte-specific markers observed in normoxia. Our study demonstrates that oxygen tension is a key factor for chondrogenesis and suggests that UBC-MSCs 3D-culture should begin in normoxia to obtain a more efficient chondrocyte differentiation before placing them in hypoxia for chondrocyte phenotype stabilization. UCB-MSCs are therefore a reliable source for cartilage engineering.
Collapse
|
29
|
Cui ZK, Sun JA, Baljon JJ, Fan J, Kim S, Wu BM, Aghaloo T, Lee M. Simultaneous delivery of hydrophobic small molecules and siRNA using Sterosomes to direct mesenchymal stem cell differentiation for bone repair. Acta Biomater 2017; 58:214-224. [PMID: 28578107 DOI: 10.1016/j.actbio.2017.05.057] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/22/2017] [Accepted: 05/30/2017] [Indexed: 01/15/2023]
Abstract
The use of small molecular drugs with gene manipulation offers synergistic therapeutic efficacy by targeting multiple signaling pathways for combined treatment. Stimulation of mesenchymal stem cells (MSCs) with osteoinductive small molecule phenamil combined with suppression of noggin is a promising therapeutic strategy that increases bone morphogenetic protein (BMP) signaling and bone repair. Our cationic Sterosome formulated with stearylamine (SA) and cholesterol (Chol) is an attractive co-delivery system that not only forms stable complexes with small interfering RNA (siRNA) molecules but also solubilizes hydrophobic small molecules in a single vehicle, for directing stem cell differentiation. Herein, we demonstrate the ability of SA/Chol Sterosomes to simultaneously deliver hydrophobic small molecule phenamil and noggin-directed siRNA to enhance osteogenic differentiation of MSCs both in in vitro two- and three-dimensional settings as well as in a mouse calvarial defect model. These results suggest a novel liposomal platform to simultaneously deliver therapeutic genes and small molecules for combined therapy. STATEMENT OF SIGNIFICANCE Application of phenamil, a small molecular bone morphogenetic protein (BMP) stimulator, combined with suppression of natural BMP antagonists such as noggin is a promising therapeutic strategy to enhance bone regeneration. Here, we present a novel strategy to co-deliver hydrophobic small molecule phenamil and noggin-targeted siRNA via cationic Sterosomes formed with stearylamine (SA) and high content of cholesterol (Chol) to enhance osteogenesis and bone repair. SA/Chol Sterosomes demonstrated high phenamil encapsulation efficiency, supported sustained release of encapsulated drugs, and significantly reduced drug dose requirements to induce osteogenic differentiation of mesenchymal stem cells (MSCs). Simultaneous deliver of phenamil and noggin siRNA in a single vehicle synergistically enhanced MSC osteogenesis and calvarial bone repair. This study suggests a new non-phospholipid liposomal formulation to simultaneously deliver small molecules and therapeutic genes for combined treatment.
Collapse
|
30
|
Tajima S, Tabata Y. Preparation of cell aggregates incorporating gelatin hydrogel microspheres containing bone morphogenic protein-2 with different degradabilities. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 29:775-792. [DOI: 10.1080/09205063.2017.1358547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Shuhei Tajima
- Department of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Amirpour N, Razavi S, Esfandiari E, Hashemibeni B, Kazemi M, Salehi H. Hanging drop culture enhances differentiation of human adipose-derived stem cells into anterior neuroectodermal cells using small molecules. Int J Dev Neurosci 2017; 59:21-30. [PMID: 28285945 DOI: 10.1016/j.ijdevneu.2017.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/04/2017] [Accepted: 03/05/2017] [Indexed: 01/26/2023] Open
Abstract
Inspired by in vivo developmental process, several studies were conducted to design a protocol for differentiating of mesenchymal stem cells into neural cells in vitro. Human adipose-derived stem cells (hADSCs) as mesenchymal stem cells are a promising source for this purpose. At current study, we applied a defined neural induction medium by using small molecules for direct differentiation of hADSCs into anterior neuroectodermal cells. Anterior neuroectodermal differentiation of hADSCs was performed by hanging drop and monolayer protocols. At these methods, three small molecules were used to suppress the BMP, Nodal, and Wnt signaling pathways in order to obtain anterior neuroectodermal (eye field) cells from hADSCs. After two and three weeks of induction, the differentiated cells with neural morphology expressed anterior neuroectodermal markers such as OTX2, SIX3, β-TUB III and PAX6. The protein expression of such markers was confirmed by real time, RT-PCR and immunocytochemistry methods According to our data, it seems that the hanging drop method is a proper approach for neuroectodermal induction of hADSCs. Considering wide availability and immunosuppressive properties of hADSCs, these cells may open a way for autologous cell therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Noushin Amirpour
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batoul Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
32
|
Salehi H, Amirpour N, Niapour A, Razavi S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev Rep 2016; 12:26-41. [PMID: 26490462 DOI: 10.1007/s12015-015-9631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide interest in application of adult stem cells due to easy to obtain with a minimal patient discomfort, capable of producing cell numbers in large quantities and their immunocompatible properties without restriction by ethical concerns. Among these stem cells, multipotent mesenchymal stem cells (MSCs) from human adipose tissue are considered as an ideal source for various regenerative medicine. In spite of mesodermal origin of human adipose-derived stem cells (hADSCs), these cells have differentiation potential toward mesodermal and non-mesodermal lineages. Up to now, several studies have shown that hADSCs can undergo transdifferentiation and produce cells outside of their lineage, especially into neural cells when they are transferred to a specific cell environment. The purpose of this literature review is to provide an overview of the existing state of knowledge of the differentiation potential of hADSCs, specifically their ability to give rise to neuronal cells. The following review discusses different protocols considered for differentiation of hADSCs to neural cells, the neural markers that are used in each procedure and possible mechanisms that are involved in this differentiation.
Collapse
|
33
|
Fan J, Guo M, Im CS, Pi-Anfruns J, Cui ZK, Kim S, Wu BM, Aghaloo TL, Lee M. Enhanced Mandibular Bone Repair by Combined Treatment of Bone Morphogenetic Protein 2 and Small-Molecule Phenamil. Tissue Eng Part A 2016; 23:195-207. [PMID: 27771997 DOI: 10.1089/ten.tea.2016.0308] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Growth factor-based therapeutics using bone morphogenetic protein 2 (BMP-2) presents a promising strategy to reconstruct craniofacial bone defects such as mandible. However, clinical applications require supraphysiological BMP doses that often increase inappropriate adipogenesis, resulting in well-documented, cyst-like bone formation. Here we reported a novel complementary strategy to enhance osteogenesis and mandibular bone repair by using small-molecule phenamil that has been shown to be a strong activator of BMP signaling. Phenamil synergistically induced osteogenic differentiation of human bone marrow mesenchymal stem cells with BMP-2 while suppressing their adipogenic differentiation induced by BMP-2 in vitro. The observed pro-osteogenic and antiadipogenic activity of phenamil was mediated by expression of tribbles homolog 3 (Trb3) that enhanced BMP-smad signaling and inhibited expression of peroxisome proliferator-activated receptor gamma (PPARγ), a master regulator of adipogenesis. The synergistic effect of BMP-2+phenamil on bone regeneration was further confirmed in a critical-sized rat mandibular bone defect by implanting polymer scaffolds designed to slowly release the therapeutic molecules. These findings indicate a new complementary osteoinductive strategy to improve clinical efficacy and safety of current BMP-based therapeutics.
Collapse
Affiliation(s)
- Jiabing Fan
- 1 Division of Advanced Prosthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Mian Guo
- 2 Department of Neurosurgery, The 2nd Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Choong Sung Im
- 1 Division of Advanced Prosthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Joan Pi-Anfruns
- 3 Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Zhong-Kai Cui
- 1 Division of Advanced Prosthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Soyon Kim
- 4 Department of Bioengineering, University of California , Los Angeles, Los Angeles, California
| | - Benjamin M Wu
- 1 Division of Advanced Prosthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California.,4 Department of Bioengineering, University of California , Los Angeles, Los Angeles, California
| | - Tara L Aghaloo
- 3 Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Min Lee
- 1 Division of Advanced Prosthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California.,4 Department of Bioengineering, University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
34
|
Tsekoura EK, K C RB, Uludag H. Biomaterials to Facilitate Delivery of RNA Agents in Bone Regeneration and Repair. ACS Biomater Sci Eng 2016; 3:1195-1206. [PMID: 33440509 DOI: 10.1021/acsbiomaterials.6b00387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bone healing after traumatic injuries or pathological diseases remains an important worldwide problem. In search of safer and more effective approaches to bone regeneration and repair, RNA-based therapeutic agents, specifically microRNAs (miRNAs) and short interfering RNA (siRNA), are beginning to be actively explored. In this review, we summarize current attempts to employ miRNAs and siRNAs in preclinical models of bone repair. We provide a summary of current limitations when attempting to utilize bioactive nucleic acids for therapeutic purposes and position the unique aspects of RNA reagents for clinical bone repair. Delivery strategies for RNA reagents are emphasized and nonviral carriers (biomaterial-based) employed to deliver such reagents are reviewed. Critical features of biomaterial carriers and various delivery technologies centered around nanoparticulate systems are highlighted. We conclude with the authors' perspectives on the future of the field, outlining main critical issues important to address as RNA reagents are explored for clinical applications.
Collapse
Affiliation(s)
- Eleni K Tsekoura
- Department of Chemical & Materials Engineering, Faculty of Engineering, ‡Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Remant Bahadur K C
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Hasan Uludag
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
35
|
Li KC, Lo SC, Sung LY, Liao YH, Chang YH, Hu YC. Improved calvarial bone repair by hASCs engineered with Cre/loxP-based baculovirus conferring prolonged BMP-2 and MiR-148b co-expression. J Tissue Eng Regen Med 2016; 11:3068-3077. [PMID: 27687795 DOI: 10.1002/term.2208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/22/2016] [Accepted: 04/05/2016] [Indexed: 12/18/2022]
Abstract
Repairing large calvarial bone defects remains a challenging task. Previously, it was discovered that that miR-148b, when acting in concert with bone morphogenetic protein 2 (BMP-2), enhanced the osteogenesis of human adipose-derived stem cells (hASCs) and improved calvarial bone healing in nude mice. However, the molecular target of miR-148b remained elusive. Here it is revealed that miR-148b directly targets NOG, whose gene product (noggin) is an antagonist to BMPs and negatively regulates BMP-induced osteogenic differentiation and bone formation. A new Cre/loxP-based baculovirus system was employed to drive prolonged BMP-2 and miR-148b overexpression in hASCs, wherein the BMP-2 overexpression induced noggin expression but the concurrent miR-148b expression downregulated noggin, thus relieving the negative regulatory loop and ameliorating hASC osteogenesis without hindering hASC proliferation or triggering appreciable cytotoxicity. Implantation of the engineered hASCs coexpressing BMP-2 and miR-148b into nude mice enabled substantial repair of critical-size calvarial bone defects (4 mm diameter) at 12 weeks post-transplantation, filling 83% of the defect area, 75% of bone volume and restoring the bone density to 89% of the original bone density. Such superior healing effects indicate the potential of the Cre/loxP-based baculovirus-mediated BMP-2/miR-148b expression for calvarial bone repair. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Chun Lo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Yu Sung
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hsin Liao
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Han Chang
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Orthopaedic, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
36
|
Lu S, Wang J, Ye J, Zou Y, Zhu Y, Wei Q, Wang X, Tang S, Liu H, Fan J, Zhang F, Farina EM, Mohammed MM, Song D, Liao J, Huang J, Guo D, Lu M, Liu F, Liu J, Li L, Ma C, Hu X, Lee MJ, Reid RR, Ameer GA, Zhou D, He T. Bone morphogenetic protein 9 (BMP9) induces effective bone formation from reversibly immortalized multipotent adipose-derived (iMAD) mesenchymal stem cells. Am J Transl Res 2016; 8:3710-3730. [PMID: 27725853 PMCID: PMC5040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/25/2016] [Indexed: 06/06/2023]
Abstract
Regenerative medicine and bone tissue engineering using mesenchymal stem cells (MSCs) hold great promise as an effective approach to bone and skeletal reconstruction. While adipose tissue harbors MSC-like progenitors, or multipotent adipose-derived cells (MADs), it is important to identify and characterize potential biological factors that can effectively induce osteogenic differentiation of MADs. To overcome the time-consuming and technically challenging process of isolating and culturing primary MADs, here we establish and characterize the reversibly immortalized mouse multipotent adipose-derived cells (iMADs). The isolated mouse primary inguinal MAD cells are reversibly immortalized via the retrovirus-mediated expression of SV40 T antigen flanked with FRT sites. The iMADs are shown to express most common MSC markers. FLP-mediated removal of SV40 T antigen effectively reduces the proliferative activity and cell survival of iMADs, indicating the immortalization is reversible. Using the highly osteogenic BMP9, we find that the iMADs are highly responsive to BMP9 stimulation, express multiple lineage regulators, and undergo osteogenic differentiation in vitro upon BMP9 stimulation. Furthermore, we demonstrate that BMP9-stimulated iMADs form robust ectopic bone with a thermoresponsive biodegradable scaffold material. Collectively, our results demonstrate that the reversibly immortalized iMADs exhibit the characteristics of multipotent MSCs and are highly responsive to BMP9-induced osteogenic differentiation. Thus, the iMADs should provide a valuable resource for the study of MAD biology, which would ultimately enable us to develop novel and efficacious strategies for MAD-based bone tissue engineering.
Collapse
Affiliation(s)
- Shun Lu
- Shandong Provincial Orthopaedics Hospital, The Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Jing Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Department of Biomedical Engineering, School of Bioengineering, Chongqing UniversityChongqing 400044, China
| | - Yulong Zou
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Yunxiao Zhu
- Department of Biomedical Engineering and Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern UniversityEvanston, IL 60208, USA
| | - Qiang Wei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Xin Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Departments of Surgery, Conservative Dentistry and Endodontics, West China Hospital and West China School of Stomatology, Sichuan UniversityChengdu 610041, China
| | - Shengli Tang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Department of General Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan UniversityWuhan 430071, China
| | - Hao Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Fugui Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Evan M Farina
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Maryam M Mohammed
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Dongzhe Song
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Departments of Surgery, Conservative Dentistry and Endodontics, West China Hospital and West China School of Stomatology, Sichuan UniversityChengdu 610041, China
| | - Junyi Liao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Jiayi Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Dan Guo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Minpeng Lu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Feng Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Jianxiang Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science & TechnologyWuhan 430022, China
| | - Li Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Department of Biomedical Engineering, School of Bioengineering, Chongqing UniversityChongqing 400044, China
| | - Chao Ma
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Department of General Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan UniversityWuhan 430071, China
| | - Xue Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Russell R Reid
- Section of Plastic Surgery, Department of Surgery, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering and Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern UniversityEvanston, IL 60208, USA
- Department of Surgery, Feinberg School of MedicineChicago, IL 60616, USA
| | - Dongsheng Zhou
- Shandong Provincial Orthopaedics Hospital, The Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Tongchuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, The Affiliated Hospitals of Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
37
|
Fan J, Im CS, Guo M, Cui ZK, Fartash A, Kim S, Patel N, Bezouglaia O, Wu BM, Wang CY, Aghaloo TL, Lee M. Enhanced Osteogenesis of Adipose-Derived Stem Cells by Regulating Bone Morphogenetic Protein Signaling Antagonists and Agonists. Stem Cells Transl Med 2016; 5:539-51. [PMID: 26956209 PMCID: PMC4798741 DOI: 10.5966/sctm.2015-0249] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Although adipose-derived stem cells (ASCs) are an attractive cell source for bone tissue engineering, direct use of ASCs alone has had limited success in the treatment of large bone defects. Although bone morphogenetic proteins (BMPs) are believed to be the most potent osteoinductive factors to promote osteogenic differentiation of ASCs, their clinical applications require supraphysiological dosage, leading to high medical burden and adverse side effects. In the present study, we demonstrated an alternative approach that can effectively complement the BMP activity to maximize the osteogenesis of ASCs without exogenous application of BMPs by regulating levels of antagonists and agonists to BMP signaling. Treatment of ASCs with the amiloride derivative phenamil, a positive regulator of BMP signaling, combined with gene manipulation to suppress the BMP antagonist noggin, significantly enhanced osteogenic differentiation of ASCs through increased BMP-Smad signaling in vitro. Furthermore, the combination approach of noggin suppression and phenamil stimulation enhanced the BMP signaling and bone repair in a mouse calvarial defect model by adding noggin knockdown ASCs to apatite-coated poly(lactic-coglycolic acid) scaffolds loaded with phenamil. These results suggest novel complementary osteoinductive strategies that could maximize activity of the BMP pathway in ASC bone repair while reducing potential adverse effects of current BMP-based therapeutics. SIGNIFICANCE Although stem cell-based tissue engineering strategy offers a promising alternative to repair damaged bone, direct use of stem cells alone is not adequate for challenging healing environments such as in large bone defects. This study demonstrates a novel strategy to maximize bone formation pathways in osteogenic differentiation of mesenchymal stem cells and functional bone formation by combining gene manipulation with a small molecule activator toward osteogenesis. The findings indicate promising stem cell-based therapy for treating bone defects that can effectively complement or replace current osteoinductive therapeutics.
Collapse
Affiliation(s)
- Jiabing Fan
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Choong Sung Im
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Mian Guo
- Department of Neurosurgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilonjiang, People's Republic of China
| | - Zhong-Kai Cui
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Armita Fartash
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Soyon Kim
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Nikhil Patel
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Olga Bezouglaia
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Benjamin M Wu
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Tara L Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Min Lee
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
38
|
Viswanathan G, Hsu YH, Voon SH, Imae T, Siriviriyanun A, Lee HB, Kiew LV, Chung LY, Yusa SI. A Comparative Study of Cellular Uptake and Subcellular Localization of Doxorubicin Loaded in Self-Assemblies of Amphiphilic Copolymers with Pendant Dendron by MDA-MB-231 Human Breast Cancer Cells. Macromol Biosci 2016; 16:882-95. [PMID: 26900760 DOI: 10.1002/mabi.201500435] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/07/2016] [Indexed: 12/19/2022]
Abstract
Previously synthesized amphiphilic diblock copolymers with pendant dendron moieties have been investigated for their potential use as drug carriers to improve the delivery of an anticancer drug to human breast cancer cells. Diblock copolymer (P71 D3 )-based micelles effectively encapsulate the doxorubicin (DOX) with a high drug-loading capacity (≈95%, 104 DOX molecules per micelle), which is approximately double the amount of drug loaded into the diblock copolymer (P296 D1 ) vesicles. DOX released from the resultant P71 D3 /DOX micelles is approximately 1.3-fold more abundant, at a tumoral acidic pH of 5.5 compared with a pH of 7.4. The P71 D3 /DOX micelles also enhance drug potency in breast cancer MDA-MB-231 cells due to their higher intracellular uptake, by approximately twofold, compared with the vesicular nanocarrier, and free DOX. Micellar nanocarriers are taken up by lysosomes via energy-dependent processes, followed by the release of DOX into the cytoplasm and subsequent translocation into the nucleus, where it exert its cytotoxic effect.
Collapse
Affiliation(s)
- Geetha Viswanathan
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yu-Hsuan Hsu
- Department of Chemical Engineering, National Taiwan University of Science and Technology, 43 Section 4, Keelung Road, Taipei, 10607, Taiwan
| | - Siew Hui Voon
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Toyoko Imae
- Department of Chemical Engineering, National Taiwan University of Science and Technology, 43 Section 4, Keelung Road, Taipei, 10607, Taiwan.,Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, 43 Section 4, Keelung Road, Taipei, 10607, Taiwan
| | - Ampornphan Siriviriyanun
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, 43 Section 4, Keelung Road, Taipei, 10607, Taiwan
| | - Hong Boon Lee
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lip Yong Chung
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Shin-Ichi Yusa
- Department of Materials Science and Chemistry, University of Hyogo, 2167 Shosha, Himeji, Hyogo, 671-2280, Japan
| |
Collapse
|
39
|
Escobar CH, Chaparro O. Xeno-Free Extraction, Culture, and Cryopreservation of Human Adipose-Derived Mesenchymal Stem Cells. Stem Cells Transl Med 2016; 5:358-65. [PMID: 26838269 DOI: 10.5966/sctm.2015-0094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 12/04/2015] [Indexed: 01/27/2023] Open
Abstract
Molecules of animal or bacterial origin, which pose a risk for zoonoses or immune rejection, are commonly used for extraction, culture, and cryopreservation of mesenchymal stem cells. There is no sequential and orderly protocol for producing human adipose-derived stem cells (hASCs) under xeno-free conditions. After standardizing a human platelet lysate (hPL) production protocol, four human adipose tissue samples were processed through explants with fetal bovine serum (FBS)-supplemented or hPL-supplemented media for extracting the adipose-derived stem cells. The cells were cultivated in cell culture medium + hPL (5%) or FBS (10%). The cellular replication rate, immunophenotype, and differentiation potential were evaluated at fourth passage. Cellular viability was evaluated before and after cryopreservation of the cells, with an hPL-based solution compared with an FBS-based solution. The explants cultured in hPL-supplemented media showed earlier and faster hASC proliferation than did those supplemented with FBS. Likewise, cells grown in hPL-supplemented media showed a greater proliferation rate, without losing the immunophenotype. Osteogenic differentiation of xeno-free hASC was higher than the hASC produced in standard conditions. However, adipogenic differentiation was reduced in xeno-free hASC. Finally, the cells cryopreserved in an hPL-based solution showed a higher cellular viability than the cells cryopreserved in an FBS-based. In conclusion, we have developed a complete xeno-free protocol for extracting, culturing, and cryopreserving hASCs that can be safely implemented in clinical studies.
Collapse
Affiliation(s)
- Carlos Hugo Escobar
- Basic Science Department, Medicine School, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia Physiology Department, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Orlando Chaparro
- Physiology Department, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
40
|
Chan SCW, Tekari A, Benneker LM, Heini PF, Gantenbein B. Osteogenic differentiation of bone marrow stromal cells is hindered by the presence of intervertebral disc cells. Arthritis Res Ther 2015; 18:29. [PMID: 26809343 PMCID: PMC4727301 DOI: 10.1186/s13075-015-0900-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Background Clinical observations indicate that the presence of nucleus pulposus (NP) tissue during spinal fusion hinders the rate of disc ossification. While the underlying mechanism remains unknown, this observation could be due to incomplete removal of NP cells (NPCs) that secrete factors preventing disc calcification, such as bone morphogenetic protein (BMP) antagonists including noggin and members of the DAN (differential screening selected gene aberrative in neuroblastoma) family. Methods Monolayer human bone marrow-derived mesenchymal stem cells (MSCs) were cocultured withNPCs and annulus fibrosus cells (AFCs) embedded in alginate for 21 days. At the end of coculture, MSCs were stained for mineral deposition by alizarin red, and relative expression of bone-related genes [Runt-related transcription factor 2, (RUNX2), Osteopontin (OPN), and Alkaline phosphatase (ALP)] and ALP activity were analyzed. Relative expression of three BMP antagonists, chordin (CHRD), gremlin (GREM1), and noggin (NOG), was determined in primary human NPCs and AFCs. These cells were also stained for Gremlin and Noggin by immunocytochemistry. Results Alizarin red staining showed that MSC osteogenesis in monolayer cultures was inhibited by coculture with NPCs or AFCs. ALP activity and RT-PCR analyses confirmed these results and demonstrated inhibition of osteogenesis of MSC in the presence of disc cells. NOG was significantly up-regulated in MSCs after coculture. Relative gene expression of intervertebral disc (IVD) cells showed higher expression of GREM1 in NPCs than in AFCs. Conclusions We show that primary IVD cells inhibit osteogenesis of MSCs. BMP inhibitors NOG, GREM1 and CHRD were expressed in IVD cells. GREM1 appears to be differentially expressed in NPCs and AFCs. Our results have implications for the design and development of treatments for non-union in spinal fusion.
Collapse
Affiliation(s)
- Samantha C W Chan
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland. .,Biointerfaces, EMPA, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St Gallen, CH-9014, Switzerland.
| | - Adel Tekari
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland.
| | - Lorin M Benneker
- Department for Orthopedic Surgery and Traumatology, Inselspital, University of Bern, Freiburgstrasse 4, Bern, CH-3010, Switzerland. .,AOSpine Research Network, Stettbachstrasse 6, Dübendorf, CH-8600, Switzerland.
| | - Paul F Heini
- Orthopedic Department, Sonnenhof Clinic, Buchserstrasse 30, Bern, CH-3006, Switzerland.
| | - Benjamin Gantenbein
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland. .,AOSpine Research Network, Stettbachstrasse 6, Dübendorf, CH-8600, Switzerland.
| |
Collapse
|
41
|
Cordeiro IR, Lopes DV, Abreu JG, Carneiro K, Rossi MID, Brito JM. Chick embryo xenograft model reveals a novel perineural niche for human adipose-derived stromal cells. Biol Open 2015; 4:1180-93. [PMID: 26319582 PMCID: PMC4582113 DOI: 10.1242/bio.010256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human adipose-derived stromal cells (hADSC) are a heterogeneous cell population that contains adult multipotent stem cells. Although it is well established that hADSC have skeletal potential in vivo in adult organisms, in vitro assays suggest further differentiation capacity, such as into glia. Thus, we propose that grafting hADSC into the embryo can provide them with a much more instructive microenvironment, allowing the human cells to adopt diverse fates or niches. Here, hADSC spheroids were grafted into either the presumptive presomitic mesoderm or the first branchial arch (BA1) regions of chick embryos. Cells were identified without previous manipulations via human-specific Alu probes, which allows efficient long-term tracing of heterogeneous primary cultures. When grafted into the trunk, in contrast to previous studies, hADSC were not found in chondrogenic or osteogenic territories up to E8. Surprisingly, 82.5% of the hADSC were associated with HNK1+ tissues, such as peripheral nerves. Human skin fibroblasts showed a smaller tropism for nerves. In line with other studies, hADSC also adopted perivascular locations. When grafted into the presumptive BA1, 74.6% of the cells were in the outflow tract, the final goal of cardiac neural crest cells, and were also associated with peripheral nerves. This is the first study showing that hADSC could adopt a perineural niche in vivo and were able to recognize cues for neural crest cell migration of the host. Therefore, we propose that xenografts of human cells into chick embryos can reveal novel behaviors of heterogeneous cell populations, such as response to migration cues.
Collapse
Affiliation(s)
- Ingrid R Cordeiro
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Daiana V Lopes
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - José G Abreu
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Katia Carneiro
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Maria I D Rossi
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - José M Brito
- Morphological Sciences Program, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| |
Collapse
|
42
|
Cui ZK, Fan J, Kim S, Bezouglaia O, Fartash A, Wu BM, Aghaloo T, Lee M. Delivery of siRNA via cationic Sterosomes to enhance osteogenic differentiation of mesenchymal stem cells. J Control Release 2015; 217:42-52. [PMID: 26302903 DOI: 10.1016/j.jconrel.2015.08.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/15/2015] [Indexed: 01/09/2023]
Abstract
Noggin is a specific antagonist of bone morphogenetic proteins (BMPs) that can prevent the interaction of BMPs with their receptors. RNA interfering molecules have been used to downregulate noggin expression and thereby stimulate BMP signaling and osteogenesis. Cationic liposomes are considered one of the most efficient non-viral systems for gene delivery. In the past decade, non-phospholipid liposomes (Sterosomes) formulated with single-chain amphiphiles and high content of sterols have been developed. In particular, Sterosomes composed of stearylamine (SA) and cholesterol (Chol) display distinct properties compared with traditional phospholipid liposomes, including increased positive surface charges and enhanced particle stability. Herein, we report SA/Chol Sterosome and small interfering RNA (siRNA) complexes that significantly enhanced cellular uptake and gene knockdown efficiencies in adipose derived mesenchymal stem cells with minimal cytotoxicity compared with commercially available lipofectamine 2000. Furthermore, we confirmed osteogenic efficacy of these Sterosomes loaded with noggin siRNA in in vitro two- and three-dimensional settings as well as in a mouse calvarial defect model. The delivery of siRNA via novel SA/Chol Sterosomes presents a powerful method for efficient gene knockdown. These distinct nanoparticles may present a promising alternative approach for gene delivery.
Collapse
Affiliation(s)
- Zhong-Kai Cui
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States
| | - Soyon Kim
- Department of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Olga Bezouglaia
- Division of Diagnostic and Surgical Sciences, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States
| | - Armita Fartash
- Division of Diagnostic and Surgical Sciences, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States
| | - Benjamin M Wu
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States; Department of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States
| | - Min Lee
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, United States; Department of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| |
Collapse
|
43
|
Abstract
Transcription factors (TFs) are multidomain proteins that play a critical role in orchestrating stem cell differentiation, but several limitations hinder the full potential of TF-based gene regulation. Here we report a unique strategy to emulate TFs and differentiate stem cells in a nonviral approach using an artificial, nanoparticle-based transcription factor called NanoScript. The NanoScript platform consists of a gold nanoparticle functionalized with small molecules that mimic the various domains of TFs. As a result, NanoScript mimics the function and structure of TF proteins. Specifically, NanoScript was designed to regulate muscle cell differentiation by targeting myogenic regulatory factors (MRFs), which play an important role in inducing myogenesis. This NanoScript-MRF is stable in physiological environments, localizes within the nucleus, induces differentiation of adipose-derived mesenchymal stem cells into mature muscle cells in 7 days, and is naturally excreted from induced muscle cells. As such, NanoScript represents a safe and powerful tool for applications requiring gene manipulation.
Collapse
Affiliation(s)
- Sahishnu Patel
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Perry T. Yin
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hiroshi Sugiyama
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8501, Japan
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
44
|
Fan J, Im CS, Cui ZK, Guo M, Bezouglaia O, Fartash A, Lee JY, Nguyen J, Wu BM, Aghaloo T, Lee M. Delivery of Phenamil Enhances BMP-2-Induced Osteogenic Differentiation of Adipose-Derived Stem Cells and Bone Formation in Calvarial Defects. Tissue Eng Part A 2015; 21:2053-65. [PMID: 25869476 DOI: 10.1089/ten.tea.2014.0489] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) have been widely used for bone repair in the craniofacial region. However, its high dose requirement in clinical applications revealed adverse effects and inefficient bone formation, along with high cost. Here, we report a novel osteoinductive strategy to effectively complement the osteogenic activity of BMP-2 using phenamil, a small molecule that can induce osteogenic differentiation via stimulation of BMP signaling. Treatment of adipose-derived stem cells (ASCs) with BMP-2 in combination with phenamil significantly promoted the in vitro osteogenic differentiation of ASCs. The efficacy of the combination strategy of phenamil+BMP-2 was further confirmed in a mouse calvarial defect model using scaffolds consisting of poly(lactic-co-glycolic acid) and apatite layer on their surfaces designed to slowly release phenamil and BMP-2. Six weeks after implantation, the scaffolds treated with phenamil+BMP-2 significantly promoted mouse calvarial regeneration as demonstrated by micro-computerized tomography and histology, compared with the groups treated with phenamil or BMP-2 alone. Moreover, the combination treatment reduced the BMP-2 dose without compromising calvarial healing efficacy. These results suggest promising complementary therapeutic strategies for bone repair in more efficient and cost-effective manners.
Collapse
Affiliation(s)
- Jiabing Fan
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Choong Sung Im
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Zhong-Kai Cui
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Mian Guo
- 2 Department of Neurosurgery, the 2nd Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Olga Bezouglaia
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Armita Fartash
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Ju-Yeon Lee
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - John Nguyen
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Benjamin M Wu
- 4 Department of Bioengineering, University of California , Los Angeles, California
| | - Tara Aghaloo
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Min Lee
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California.,4 Department of Bioengineering, University of California , Los Angeles, California
| |
Collapse
|
45
|
Aryal R, Chen XP, Fang C, Hu YC. Bone morphogenetic protein-2 and vascular endothelial growth factor in bone tissue regeneration: new insight and perspectives. Orthop Surg 2015; 6:171-8. [PMID: 25179350 DOI: 10.1111/os.12112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/18/2014] [Indexed: 12/20/2022] Open
Abstract
The study of bone tissue regeneration in orthopaedic diseases has stimulated great interest among bone tissue engineering specialists and orthopaedic surgeons. Combinations of biomaterials, growth factors and stem cells for repairing bone have been much studied and researched, yet remain a challenge for both scientists and clinicians pursuing regenerative medicine. The purpose of this review was to elucidate the role of sequential release of bone morphogenetic protein-2 and vascular endothelial growth factor in producing better outcomes in the field of bone tissue regeneration.
Collapse
|
46
|
Choi B, Kim S, Fan J, Kowalski T, Petrigliano F, Evseenko D, Lee M. Covalently conjugated transforming growth factor-β1 in modular chitosan hydrogels for the effective treatment of articular cartilage defects. Biomater Sci 2015. [PMID: 26222593 DOI: 10.1039/c4bm00431k] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Approaches to control precisely growth factor presentation to a tissue defect in a sustained fashion are of increasing interest for a number of complex tissue engineering applications. Although transforming growth factor beta-1 (TGF-β1) plays a key role in promoting chondrogenesis, the therapeutic use of TGF-β1 is limited by its inherent protein instability, requiring high amounts of the protein that can cause adverse side effects with inefficient cartilage formation. In this work, we have developed strategies to stabilize TGF-β1 signaling in the injectable, visible blue light inducible chitosan (MeGC) hydrogel system for specific use in cartilage regeneration. We successfully modulated delivery of TGF-β1 with reduced burst release in a complex biological environment of serum and cells by covalently conjugating the protein to MeGC hydrogels with preserving type II collagen, one of the major cartilaginous extracellular matrix (ECM) components. The hydrogel system supported cellular condensation and deposition of cartilaginous ECM by encapsulating adipose derived stem cells in vitro. We confirmed further the ability of these TGF-β1 functionalized hydrogel systems to promote cartilage regeneration in challenging healing environments such as in a rat partial-thickness chondral defect model which present a limited source of subchondral bone marrow elements. These results suggest a new injectable delivery modality of therapeutic agents to improve clinical cartilage repair.
Collapse
Affiliation(s)
- Bogyu Choi
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Kim J, Lin B, Kim S, Choi B, Evseenko D, Lee M. TGF-β1 conjugated chitosan collagen hydrogels induce chondrogenic differentiation of human synovium-derived stem cells. J Biol Eng 2015; 9:1. [PMID: 25745515 PMCID: PMC4350967 DOI: 10.1186/1754-1611-9-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/24/2014] [Indexed: 12/27/2022] Open
Abstract
Background Unlike bone tissue, articular cartilage regeneration has not been very successful and has many challenges ahead. We have previously developed injectable hydrogels using photopolymerizable chitosan (MeGC) that supported growth of chondrocytes. In this study, we demonstrate a biofunctional hydrogel for specific use in cartilage regeneration by conjugating transforming growth factor-β1 (TGF-β1), a well-documented chondrogenic factor, to MeGC hydrogels impregnating type II collagen (Col II), one of the major cartilaginous extracellular matrix (ECM) components. Results TGF-β1 was delivered from MeGC hydrogels in a controlled manner with reduced burst release by chemically conjugating the protein to MeGC. The hydrogel system did not compromise viability of encapsulated human synovium-derived mesenchymal stem cells (hSMSCs). Col II impregnation and TGF-β1 delivery significantly enhanced cellular aggregation and deposition of cartilaginous ECM by the encapsulated cells, compared with pure MeGC hydrogels. Conclusions This study demonstrates successful engineering of a biofunctional hydrogel with a specific microenvironment tailored to promote chondrogenesis. This hydrogel system can provide promising efficacious therapeutics in the treatment of cartilage defects. Electronic supplementary material The online version of this article (doi:10.1186/1754-1611-9-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, 339-701 South Korea
| | - Brian Lin
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Soyon Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Bogyu Choi
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095 USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA ; Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
48
|
Choi B, Cui ZK, Kim S, Fan J, Wu BM, Lee M. Glutamine-chitosan modified calcium phosphate nanoparticles for efficient siRNA delivery and osteogenic differentiation. J Mater Chem B 2015; 3:6448-6455. [PMID: 26413302 DOI: 10.1039/c5tb00843c] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
RNA interference (RNAi)-based therapy using small interfering RNA (siRNA) exhibits great potential to treat diseases. Although calcium phosphate (CaP)-based systems are attractive options to deliver nucleic acids due to their good biocompatibility and high affinity with nucleic acids, they are limited by uncontrollable particle formation and inconsistent transfection efficiencies. In this study, we developed a stable CaP nanocarrier system with enhanced intracellular uptake by adding highly cationic, glutamine-conjugated oligochitosan (Gln-OChi). CaP nanoparticles coated with Gln-OChi (CaP/Gln-OChi) significantly enhanced gene transfection and knockdown efficiency in both immortalized cell line (HeLa) and primary mesenchymal stem cells (MSCs) with minimal cytotoxicity. The osteogenic bioactivity of siRNA-loaded CaP/Gln-OChi particles was further confirmed in three-dimensional environments by using photocrosslinkable chitosan hydrogels encapsulating MSCs and particles loaded with siRNA targeting noggin, a bone morphogenetic protein antagonist. These findings suggest that our CaP/Gln-OChi nanocarrier provides an efficient and safe gene delivery system for therapeutic applications.
Collapse
Affiliation(s)
- Bogyu Choi
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Zhong-Kai Cui
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Soyon Kim
- Department of Bioengineering, University of California Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, USA
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Benjamin M Wu
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA ; Department of Bioengineering, University of California Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA ; Department of Bioengineering, University of California Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, USA
| |
Collapse
|
49
|
Qian D, Bai B, Yan G, Zhang S, Liu Q, Chen Y, Tan X, Zeng Y. Construction of doxycycline-mediated BMP-2 transgene combining with APA microcapsules for bone repair. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 44:270-6. [PMID: 25092431 DOI: 10.3109/21691401.2014.942458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Dongyang Qian
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Bo Bai
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Guangbin Yan
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Shujiang Zhang
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Qi Liu
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Yi Chen
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Xiaobo Tan
- a Department of Orthopaedics , the First Affiliated Hospital, Guangzhou Medical University , Guangzhou , P. R. China
| | - Yanjun Zeng
- b Biomechanics & Medical Information Institute, Beijing University of Technology , Beijing , P. R. China
| |
Collapse
|
50
|
Fan J, Park H, Lee MK, Bezouglaia O, Fartash A, Kim J, Aghaloo T, Lee M. Adipose-derived stem cells and BMP-2 delivery in chitosan-based 3D constructs to enhance bone regeneration in a rat mandibular defect model. Tissue Eng Part A 2014; 20:2169-79. [PMID: 24524819 PMCID: PMC4137352 DOI: 10.1089/ten.tea.2013.0523] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/10/2014] [Indexed: 12/29/2022] Open
Abstract
Reconstructing segmental mandiblular defects remains a challenge in the clinic. Tissue engineering strategies provide an alternative option to resolve this problem. The objective of the present study was to determine the effects of adipose-derived stem cells (ASCs) and bone morphogenetic proteins-2 (BMP-2) in three-dimensional (3D) scaffolds on mandibular repair in a small animal model. Noggin expression levels in ASCs were downregulated by a lentiviral short hairpin RNA strategy to enhance ASC osteogenesis (ASCs(Nog-)). Chitosan (CH) and chondroitin sulfate (CS), natural polysaccharides, were fabricated into 3D porous scaffolds, which were further modified with apatite coatings for enhanced cellular responses and efficient delivery of BMP-2. The efficacy of 3D apatite-coated CH/CS scaffolds supplemented with ASCs(Nog-) and BMP-2 were evaluated in a rat critical-sized mandibular defect model. After 8 weeks postimplantation, the scaffolds treated with ASCs(Nog-) and BMP-2 significantly promoted rat mandibular regeneration as demonstrated by micro-computerized tomography, histology, and immunohistochemistry, compared with the groups treated with ASCs(Nog-) or BMP-2 alone. These results suggest that our combinatorial strategy of ASCs(Nog-)+BMP-2 in 3D apatite microenvironments can significantly promote mandibular regeneration, and these may provide a potential tissue engineering approach to repair large bony defects.
Collapse
Affiliation(s)
- Jiabing Fan
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Hyejin Park
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Matthew K. Lee
- Department of Head and Neck Surgery, University of California, Los Angeles, Los Angeles, California
| | - Olga Bezouglaia
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Armita Fartash
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Korea
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Min Lee
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|