1
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
2
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
3
|
Lee KG, Santos ARMP, Kang YG, Chae YJ, Shah M, Pirzada RH, Song M, Kim J, Choi S, Park Y. Efficacy Evaluation of SDF-1α-Based Polypeptides in an Acute Myocardial Infarction Model Using Structure-Based Drug Design. ACS Biomater Sci Eng 2022; 8:4486-4496. [PMID: 36178141 DOI: 10.1021/acsbiomaterials.2c00766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Stromal cell-derived factor-1 alpha (SDF-1α, CXCL12) mediates the migration of circulating cells to desired sites for tissue development, homeostasis, and regeneration and can be used to promote cardiac regeneration by recruiting stem cells. However, the use of SDF-1α in the injured heart necessitates not only higher binding affinity to its receptor, CXCR4+, but also better robustness against enzymatic degradation than other SDF-1 isoforms. Here, we conduct a screening of SDF-1α analog peptides that were designed by structure-based drug design (SBDD), a type of computer-aided drug design (CADD). We have developed in vitro and in vivo methods that enable us to estimate the effect of peptides on the migration of human mesenchymal stem cells (hMSCs) and cardiac regeneration in acute myocardial infarction (AMI)-induced animals, respectively. We demonstrate that one type of SDF-1α analog peptide, SDP-4, among the four analog peptides preselected by SBDD, is more potent than native SDF-1α for cardiac regeneration in myocardial infarction. It is interesting to note that the migratory effects of SDP-4 determined by a wound healing assay, a Transwell assay, and a 2D migration assay are comparable to those of SDF-1α. These results suggest that in vivo, as well as in vitro, screening of peptides developed by SBDD is a quintessential process to the development of a novel therapeutic compound for cardiac regeneration. Our finding also has an implication that the SDP-4 peptide is an excellent candidate for use in the regeneration of an AMI heart.
Collapse
Affiliation(s)
- Kang-Gon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Ana Rita M P Santos
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yong Guk Kang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yun Jin Chae
- R&D center, Scholar Foxtrot Co. Ltd., Seoul 02796, Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | | | - Myeongjin Song
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jongseong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.,R&D center, Scholar Foxtrot Co. Ltd., Seoul 02796, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
4
|
SDF-1 α/OPF/BP Composites Enhance the Migrating and Osteogenic Abilities of Mesenchymal Stem Cells. Stem Cells Int 2021; 2021:1938819. [PMID: 34434236 PMCID: PMC8380507 DOI: 10.1155/2021/1938819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/02/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
In situ cell recruitment is a promising regenerative medicine strategy with the purpose of tissue regeneration without stem cell transplantation. This chemotaxis-based strategy is aimed at ensuring a restorative environment through the release of chemokines that promote site-specific migration of healing cell populations. Stromal cell-derived factor-1α (SDF-1α) is a critical chemokine that can regulate the migration of mesenchymal stem cells (MSCs). Accordingly, here, SDF-1α-loaded microporous oligo[poly(ethylene glycol) fumarate]/bis[2-(methacryloyloxy)ethyl] phosphate composites (SDF-1α/OPF/BP) were engineered and probed. SDF-1α/OPF/BP composites were loaded with escalating SDF-1α concentrations, namely, 0 ng/ml, 50 ng/ml, 100 ng/ml, and 200 ng/ml, and were cocultured with MSC. Scratching assay, Transwell assay, and three-dimensional migration model were utilized to assess the migration response of MSCs. Immunofluorescence staining of Runx2 and osteopontin (OPN), ELISA assay of osteocalcin (OCN) and alkaline phosphatase (ALP), and Alizarin Red S staining were conducted to assess the osteogenesis of MSCs. All SDF-1α/OPF/BP composites engendered a release of SDF-1α (>80%) during the first four days. SDF-1α released from the composites significantly promoted migration and osteogenic differentiation of MSCs documented by upregulated expression of osteogenic-related proteins, ALP, Runx2, OCN, and OPN. SDF-1α at 100 ng/ml was optimal for enhanced migration and osteogenic proficiency. Thus, designed SDF-1α/OPF/BP composites were competent in promoting the homing and osteogenesis of MSCs and thus offer a promising bioactive scaffold candidate for on-demand bone tissue regeneration.
Collapse
|
5
|
Lin Z, Nica C, Sculean A, Asparuhova MB. Positive Effects of Three-Dimensional Collagen-Based Matrices on the Behavior of Osteoprogenitors. Front Bioeng Biotechnol 2021; 9:708830. [PMID: 34368101 PMCID: PMC8334008 DOI: 10.3389/fbioe.2021.708830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022] Open
Abstract
Recent research has demonstrated that reinforced three-dimensional (3D) collagen matrices can provide a stable scaffold for restoring the lost volume of a deficient alveolar bone. In the present study, we aimed to comparatively investigate the migratory, adhesive, proliferative, and differentiation potential of mesenchymal stromal ST2 and pre-osteoblastic MC3T3-E1 cells in response to four 3D collagen-based matrices. Dried acellular dermal matrix (DADM), hydrated acellular dermal matrix (HADM), non-crosslinked collagen matrix (NCM), and crosslinked collagen matrix (CCM) did all enhance the motility of the osteoprogenitor cells. Compared to DADM and NCM, HADM and CCM triggered stronger migratory response. While cells grown on DADM and NCM demonstrated proliferative rates comparable to control cells grown in the absence of a biomaterial, cells grown on HADM and CCM proliferated significantly faster. The pro-proliferative effects of the two matrices were supported by upregulated expression of genes regulating cell division. Increased expression of genes encoding the adhesive molecules fibronectin, vinculin, CD44 antigen, and the intracellular adhesive molecule-1 was detected in cells grown on each of the scaffolds, suggesting excellent adhesive properties of the investigated biomaterials. In contrast to genes encoding the bone matrix proteins collagen type I (Col1a1) and osteopontin (Spp1) induced by all matrices, the expression of the osteogenic differentiation markers Runx2, Alpl, Dlx5, Ibsp, Bglap2, and Phex was significantly increased in cells grown on HADM and CCM only. Short/clinically relevant pre-coating of the 3D biomaterials with enamel matrix derivative (EMD) or recombinant bone morphogenetic protein-2 (rBMP-2) significantly boosted the osteogenic differentiation of both osteoprogenitor lines on all matrices, including DADM and NCM, indicating that EMD and BMP-2 retained their biological activity after being released from the matrices. Whereas EMD triggered the expression of all osteogenesis-related genes, rBMP-2 upregulated early, intermediate, and late osteogenic differentiation markers except for Col1a1 and Spp1. Altogether, our results support favorable influence of HADM and CCM on the recruitment, growth, and osteogenic differentiation of the osteoprogenitor cell types. Furthermore, our data strongly support the biofunctionalization of the collagen-based matrices with EMD or rBMP-2 as a potential treatment modality for bone defects in the clinical practice.
Collapse
Affiliation(s)
- Zhikai Lin
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Cristina Nica
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Maria B Asparuhova
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Bern, Switzerland.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Sorooshian P, Metcalfe AD, Lali FV. In vitro modelling of disease-induced changes in the diabetic wound fibroblast. J Wound Care 2021; 30:300-303. [PMID: 33856909 DOI: 10.12968/jowc.2021.30.4.300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Fibroblasts have been shown to play an increasingly important role within diabetic wounds. While several in vitro models of diabetic wound fibroblasts have been reported, none replicate the natural progression of the disease over time, recapitulating the acquisition of the diseased phenotype. Therefore, this study aimed to establish an in vitro model of the diabetic wound fibroblast through sustained exposure of healthy dermal fibroblasts to hyperglycaemia. METHOD Primary human fibroblasts were isolated from discarded healthy skin tissue and were either exposed to normoglycaemic (control 5.5mM glucose) media or hyperglycaemic (25mM glucose) media for four weeks. Quantitative polymerase chain reaction was performed to measure the expression of inflammatory cytokines and chemokines. RESULTS In the hyperglycaemia model, stromal cell-derived factor (SDF)-1 expression remained consistently downregulated across all four weeks (p<0.01), while monocyte chemoattractant protein (MCP)-1 (p<0.001), interleukin (IL)-8 (p=0.847) and chemokine (C-X-C motif) ligand 1 (CXCL1) (p=0.872) were initially downregulated at one week followed by subsequent upregulation between 2-4 weeks. CONCLUSION This hyperglycaemia model may serve as a useful tool to characterise pathological changes in the diabetic wound fibroblast and help identify candidate therapeutic targets, such as SDF-1, that may reverse the pathology.
Collapse
Affiliation(s)
- Parviz Sorooshian
- Department of Plastic and Reconstructive Surgery, Queen Victoria Hospital NHS Trust, East Grinstead, UK
| | - Anthony D Metcalfe
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Ferdinand V Lali
- Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospital, Harrow, UK
| |
Collapse
|
7
|
Zhang H, Li X, Li J, Zhong L, Chen X, Chen S. SDF-1 mediates mesenchymal stem cell recruitment and migration via the SDF-1/CXCR4 axis in bone defect. J Bone Miner Metab 2021; 39:126-138. [PMID: 33079278 DOI: 10.1007/s00774-020-01122-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/27/2020] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Recent studies have indicated the potential of stem cell therapy in combination with cytokines to restore the bone repair via migration and homing of stem cells to the defected area. The present study aimed to investigate the mobilization and recruitment of mesenchymal stem cells (MSCs) in response to SDF-1. MATERIALS AND METHODS Herein, the knockout rat model of the bone defect (BD) was treated with the induced membrane technique. Then, wild type Wistar rats and SDF-1-knockout rats were selected for the establishment of BD-induced membrane (BD-IM) models and bone-graft (BG) models. The number of MSCs was evaluated by flow cytometry, along with the expression pattern of the SDF-1/CXCR4 axis as well as osteogenic factors was identified by RT-qPCR and Western blot analyses. Finally, the MSC migration ability was assessed by the Transwell assay. RESULTS Our data illustrated that in the induced membrane tissues, the number of MSCs among the BD-IM modeled rats was increased, whereas, a lower number was documented among BG modeled rats. Besides, we found that lentivirus-mediated over-expression of SDF-1 in BG modeled rats could activate the SDF-1/CXCR4 axis, mobilize MSCs into the defect area, and up-regulate the osteogenic proteins. CONCLUSIONS Collectively, our study speculated that up-regulation of SDF-1 promotes the mobilization and migration of MSCs through the activation of the SDF-1/CXCR4 signal pathway.
Collapse
Affiliation(s)
- Heli Zhang
- Department of Outpatient, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Xijing Li
- Department of Emergency, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Junfeng Li
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Lili Zhong
- Jilin Provincial Key Laboratory On Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Xue Chen
- Department of Orthopedics, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, People's Republic of China.
| | - Si Chen
- Department of Geriatric Medicine, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, People's Republic of China.
| |
Collapse
|
8
|
Glaeser JD, Behrens P, Stefanovic T, Salehi K, Papalamprou A, Tawackoli W, Metzger MF, Eberlein S, Nelson T, Arabi Y, Kim K, Baloh RH, Ben-David S, Cohn-Schwartz D, Ryu R, Bae HW, Gazit Z, Sheyn D. Neural crest-derived mesenchymal progenitor cells enhance cranial allograft integration. Stem Cells Transl Med 2021; 10:797-809. [PMID: 33512772 PMCID: PMC8046069 DOI: 10.1002/sctm.20-0364] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/10/2020] [Accepted: 11/09/2020] [Indexed: 01/17/2023] Open
Abstract
Replacement of lost cranial bone (partly mesodermal and partly neural crest‐derived) is challenging and includes the use of nonviable allografts. To revitalize allografts, bone marrow‐derived mesenchymal stromal cells (mesoderm‐derived BM‐MSCs) have been used with limited success. We hypothesize that coating of allografts with induced neural crest cell‐mesenchymal progenitor cells (iNCC‐MPCs) improves implant‐to‐bone integration in mouse cranial defects. Human induced pluripotent stem cells were reprogramed from dermal fibroblasts, differentiated to iNCCs and then to iNCC‐MPCs. BM‐MSCs were used as reference. Cells were labeled with luciferase (Luc2) and characterized for MSC consensus markers expression, differentiation, and risk of cellular transformation. A calvarial defect was created in non‐obese diabetic/severe combined immunodeficiency (NOD/SCID) mice and allografts were implanted, with or without cell coating. Bioluminescence imaging (BLI), microcomputed tomography (μCT), histology, immunofluorescence, and biomechanical tests were performed. Characterization of iNCC‐MPC‐Luc2 vs BM‐MSC‐Luc2 showed no difference in MSC markers expression and differentiation in vitro. In vivo, BLI indicated survival of both cell types for at least 8 weeks. At week 8, μCT analysis showed enhanced structural parameters in the iNCC‐MPC‐Luc2 group and increased bone volume in the BM‐MSC‐Luc2 group compared to controls. Histology demonstrated improved integration of iNCC‐MPC‐Luc2 allografts compared to BM‐MSC‐Luc2 group and controls. Human osteocalcin and collagen type 1 were detected at the allograft‐host interphase in cell‐seeded groups. The iNCC‐MPC‐Luc2 group also demonstrated improved biomechanical properties compared to BM‐MSC‐Luc2 implants and cell‐free controls. Our results show an improved integration of iNCC‐MPC‐Luc2‐coated allografts compared to BM‐MSC‐Luc2 and controls, suggesting the use of iNCC‐MPCs as potential cell source for cranial bone repair.
Collapse
Affiliation(s)
- Juliane D Glaeser
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Phillip Behrens
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Wafa Tawackoli
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Melodie F Metzger
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Orthopaedic Biomechanics Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Samuel Eberlein
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Trevor Nelson
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yasaman Arabi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kevin Kim
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Orthopaedic Biomechanics Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert H Baloh
- 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
| | - Shiran Ben-David
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Doron Cohn-Schwartz
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Division of Internal Medicine, Rambam Health Care Campus, Haifa, Israel
| | - Robert Ryu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hyun W Bae
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zulma Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
9
|
Augustine R, Ur Rehman SR, K S J, Hasan A. Stromal cell-derived factor loaded co-electrospun hydrophilic/hydrophobic bicomponent membranes for wound protection and healing. RSC Adv 2020; 11:572-583. [PMID: 35423060 PMCID: PMC8691117 DOI: 10.1039/d0ra04997b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic wounds are one of the key concerns for people with diabetes, frequently leading to infections and non-healing ulcers, and finally resulting in the amputation of limbs/organs. Stromal cell-derived factor 1 (SDF1) is a major chemokine that plays a significant role in tissue repair, vascularization, and wound healing. However, the long-term sustained delivery of SDF1 in a chronic wound environment is a great challenge. In order to facilitate the sustained release of SDF1 in diabetic wounds, it could be incorporated into wound-healing patches. Herein, we report the fabrication of a hydrophilic/hydrophobic bicomponent fiber-based membrane, where SDF1 was encapsulated inside hydrophilic fibers, and its applicability in wound healing. A co-electrospinning technique was employed for the fabrication of polymeric membranes where PVA and PCL form the hydrophilic and hydrophobic components, respectively. Morphological analysis of the developed membranes was conducted via scanning electron microscopy (SEM). The mechanical strength of the membranes was investigated via uniaxial tensile testing. The water uptake capacity of the membranes was also determined to understand the hydrophilicity and exudate uptake capacity of the membranes. To understand the proliferation, viability, and migration of skin-specific cells in the presence of SDF1-loaded membranes, in vitro cell culture experiments were carried out using fibroblasts, keratinocytes, and endothelial cells. The results showed the excellent porous morphology of the developed membranes with distinguishable differences in fiber diameters for the PVA and PCL fibers. The developed membranes possessed enough mechanical strength for use as wound-healing membranes. The co-electrospun membranes showed good exudate uptake capacity. The controlled and extended delivery of SDF1 from the developed membranes was observed over a prolonged period. The SDF1-loaded membranes showed enhanced cell proliferation, cell viability, and cell migration. These biocompatible and biodegradable SDF1-loaded bicomponent membranes with excellent exudate uptake capacity, and cell proliferation and cell migration properties can be exploited as a novel wound-dressing membrane aimed at chronic diabetic wounds.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University - 2713 Doha Qatar
- Biomedical Research Center, Qatar University - 2713 Doha Qatar
| | - Syed Raza Ur Rehman
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University - 2713 Doha Qatar
- Biomedical Research Center, Qatar University - 2713 Doha Qatar
| | - Joshy K S
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University - 2713 Doha Qatar
- Biomedical Research Center, Qatar University - 2713 Doha Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University - 2713 Doha Qatar
- Biomedical Research Center, Qatar University - 2713 Doha Qatar
| |
Collapse
|
10
|
Jung Y, Kim JK, Lee E, Cackowski FC, Decker AM, Krebsbach PH, Taichman RS. CXCL12γ induces human prostate and mammary gland development. Prostate 2020; 80:1145-1156. [PMID: 32659025 PMCID: PMC7491592 DOI: 10.1002/pros.24043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/11/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Epithelial stem cells (ESCs) demonstrate a capacity to maintain normal tissues homeostasis and ESCs with a deregulated behavior can contribute to cancer development. The ability to reprogram normal tissue epithelial cells into prostate or mammary stem-like cells holds great promise to help understand cell of origin and lineage plasticity in prostate and breast cancers in addition to understanding normal gland development. We previously showed that an intracellular chemokine, CXCL12γ induced cancer stem cells and neuroendocrine characteristics in both prostate and breast adenocarcinoma cell lines. However, its role in normal prostate or mammary epithelial cell fate and development remains unknown. Therefore, we sought to elucidate the functional role of CXCL12γ in the regulation of ESCs and tissue development. METHODS Prostate epithelial cells (PNT2) or mammary epithelial cells (MCF10A) with overexpressed CXCL12γ was characterized by quantitative real-time polymerase chain reaction, Western blots, and immunofluorescence for lineage marker expression, and fluorescence activated cell sorting analyses and sphere formation assays to examine stem cell surface phenotype and function. Xenotransplantation animal models were used to evaluate gland or acini formation in vivo. RESULTS Overexpression of CXCL12γ promotes the reprogramming of cells with a differentiated luminal phenotype to a nonluminal phenotype in both prostate (PNT2) and mammary (MCF10A) epithelial cells. The CXCL12γ-mediated nonluminal type cells results in an increase of epithelial stem-like phenotype including the subpopulation of EPCAMLo /CD49fHi /CD24Lo /CD44Hi cells capable of sphere formation. Critically, overexpression of CXCL12γ promotes the generation of robust gland-like structures from both prostate and mammary epithelial cells in in vivo xenograft animal models. CONCLUSIONS CXCL12γ supports the reprogramming of epithelial cells into nonluminal cell-derived stem cells, which facilitates gland development.
Collapse
Affiliation(s)
- Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Co-senior authors
| | - Jin Koo Kim
- Section of Periodontics, University of California Los Angeles School of Dentistry, Los Angeles, CA 90095, USA
| | - Eunsohl Lee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Frank C. Cackowski
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Ann M. Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Paul H. Krebsbach
- Section of Periodontics, University of California Los Angeles School of Dentistry, Los Angeles, CA 90095, USA
| | - Russell S. Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Periodontics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Co-senior authors
- Corresponding Author Russell S. Taichman D.M.D., D.M.Sc., School of Dentistry, The University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294-0007, Phone: 205-934-4720,
| |
Collapse
|
11
|
Which substances loaded onto collagen scaffolds influence oral tissue regeneration?-an overview of the last 15 years. Clin Oral Investig 2020; 24:3363-3394. [PMID: 32827278 DOI: 10.1007/s00784-020-03520-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Collagen scaffolds are widely used for guided bone or tissue regeneration. Aiming to enhance their regenerative properties, studies have loaded various substances onto these scaffolds. This review aims to provide an overview of existing literature which conducted in vitro, in vivo, and clinical testing of drug-loaded collagen scaffolds and analyze their outcome of promoting oral regeneration. MATERIALS AND METHODS PubMed, Scopus, and Ovid Medline® were systematically searched for publications from 2005 to 2019. Journal articles assessing the effect of substances on oral hard or soft tissue regeneration, while using collagen carriers, were screened and qualitatively analyzed. Studies were grouped according to their used substance type-biological medical products, pharmaceuticals, and tissue-, cell-, and matrix-derived products. RESULTS A total of 77 publications, applying 36 different substances, were included. Collagen scaffolds were demonstrating favorable adsorption behavior and release kinetics which could even be modified. BMP-2 was investigated most frequently, showing positive effects on oral tissue regeneration. BMP-9 showed comparable results at lower concentrations. Also, FGF2 enhanced bone and periodontal healing. Antibiotics improved the scaffold's anti-microbial activity and reduced the penetrability for bacteria. CONCLUSION Growth factors showed promising results for oral tissue regeneration, while other substances were investigated less frequently. Found effects of investigated substances as well as adsorption and release properties of collagen scaffolds should be considered for further investigation. CLINICAL RELEVANCE Collagen scaffolds are reliable carriers for any of the applied substances. BMP-2, BMP-9, and FGF2 showed enhanced bone and periodontal healing. Antibiotics improved anti-microbial properties of the scaffolds.
Collapse
|
12
|
Wang F, Yang G, Xiao Y, He C, Cai G, Song E, Li Y. Effect of SDF-1 with biphasic ceramic-like bone graft on the repair of rabbit radial defect. J Orthop Surg Res 2019; 14:231. [PMID: 31331380 PMCID: PMC6647097 DOI: 10.1186/s13018-019-1277-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/11/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effects of stromal cell-derived factor-1 (SDF-1) on biphasic ceramic-like biologic bone (BCBB) in vivo on the repair of large segment bone defect in rabbits. METHODS A large-segment radius defect model of the rabbits was constructed. In the experimental group, BCBB with SDF-1 sustained-release system were implanted into the bone defect site. Other three groups including normal control, autologous bone graft, and BCBB implantation without SDF-1 were set. After surgery, general observation, X-ray radiography and scoring, and tissue section staining were performed at 2, 4, 8, 12, and 24 weeks post-implantation. RESULTS By general observation, X-ray radiography and grading and tissue section staining observation, we found that the BCBB carrying SDF-1 was better than those in the group of BCBB without SDF-1 (P < 0.05). BCBB scaffold had certain bone conduction capacity, and the BCBB scaffold carrying SDF-1 had improved bone conduction ability and possessed bone induction ability. In the case of carrying SDF-1, it can be used to repair large bone defects in a shorter time than simply using BCBB, which is equivalent to the effect of autologous bone. CONCLUSION BCBB scaffold carrying SDF-1 can promote the repair effect on a large bone defect, which is equivalent to the effect of autologous bone.
Collapse
Affiliation(s)
- Fuke Wang
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - Guiran Yang
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - Yu Xiao
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - Chuan He
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - Guofeng Cai
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - En Song
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, No. 4 Building, Xichang Road, Wuhua District, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
13
|
Aryal YP, Neupane S, Adhikari N, An C, Ha J, Kwon T, Yamamoto H, Jung J, Park E, Kim J, Cho S, Sohn W, Lee Y, Chae H, Kim H, Kim J. An endoplasmic reticulum stress regulator,
Tmbim6
, modulates secretory stage of mice molar. J Cell Physiol 2019; 234:20354-20365. [DOI: 10.1002/jcp.28635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Yam Prasad Aryal
- Department of Biochemistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Sanjiv Neupane
- Department of Biochemistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Nirpesh Adhikari
- Department of Biochemistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Chang‐Hyeon An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Jung‐Hong Ha
- Department of Conservative Dentistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Tae‐Yub Kwon
- Department of Dental Biomaterials, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Hitoshi Yamamoto
- Department of Histology and Developmental Biology Tokyo Dental College Tokyo Japan
| | - Jae‐Kwang Jung
- Department of Oral Medicine, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Eui‐Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Ji‐Youn Kim
- Department of Dental Hygiene Gachon University Incheon Korea
| | - Sung‐Won Cho
- Division in Anatomy and Developmental Biology, Department of Oral Biology Yonsei University College of Dentistry Seoul Korea
| | - Wern‐Joo Sohn
- Pre‐Major of Cosmetics and Pharmaceutics Daegu Haany University Gyeongsan Korea
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| | - Han‐Jung Chae
- Department of Pharmacology and New Drug Development Institute Chonbuk National University Jeonju Korea
| | - Hyung‐Ryong Kim
- Institute of Tissue Regeneration Engineering (ITREN) Dankook University Cheonan Korea
| | - Jae‐Young Kim
- Department of Biochemistry, School of Dentistry, IHBR Kyungpook National University Daegu Korea
| |
Collapse
|
14
|
Reconstruction of a Calvarial Wound Complicated by Infection: Comparing the Effects of Biopatterned Bone Morphogenetic Protein 2 and Vascular Endothelial Growth Factor. J Craniofac Surg 2019; 30:260-264. [PMID: 30339591 DOI: 10.1097/scs.0000000000004779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) bioprinted on biological matrix induces osseous regeneration in large calvarial defects in rabbits, both uncomplicated and scarred. Healing in unfavorable defects scarred from previous infection is decreased due in part to the lack of vascularity. This impedes the access of mesenchymal stem cells, key to osseous regeneration and the efficacy of BMP2, to the wound bed. The authors hypothesized that bioprinted vascular endothelial growth factor (VEGF) would augment the osseous regeneration achieved with low dose biopatterned BMP2 alone. Thirteen New Zealand white rabbits underwent subtotal calvariectomy using a dental cutting burr. Care was taken to preserve the underlying dura. A 15 mm × 15 mm flap of bone was cut away and incubated in a 1 × 108 cfu/mL planktonic solution of S aureus before reimplantation. After 2 weeks of subsequent infection the flap was removed and the surgical wound debrided followed by 10 days of antibiotic treatment. On postoperative day 42 the calvarial defects were treated with acellular dermal matrix bioprinted with nothing (control), VEGF, BMP2, BMP2/VEGF combined. Bone growth was analyzed with serial CT and postmortem histology. Defects treated with BMP2 (BMP2 alone and BMP2/VEGF combination) showed significantly greater healing than control and VEGF treated defect (P < 0.5). Vascular endothelial growth factor treated defect demonstrated less healing than control and VEGF/BMP2 combination treatments achieved less healing than BMP2 alone though these differences were nonsignificant. Low dose BMP2-patterned acellular dermal matrix improves healing of scarred calvarial defects. Vascular endothelial growth factor at the doses applied in this study failed to increase healing.
Collapse
|
15
|
Xing F, Li L, Liu M, Duan X, Long Y, Xiang Z. [The application and research progress of in-situ tissue engineering technology in bone and cartilage repair]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:1358-1364. [PMID: 30215487 DOI: 10.7507/1002-1892.201712118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective To review the application and research progress of
in-situ tissue engineering technology in bone and cartilage repair. Methods The original articles about
in-situ tissue engineering technology in bone and cartilage repair were extensively reviewed and analyzed. Results In-situ tissue engineering have been shown to be effective in repairing bone defects and cartilage defects, but biological mechanisms are inadequate. At present, most of researches are mainly focused on animal experiments, and the effect of clinical repair need to be further studied. Conclusion In-situ tissue engineering technology has wide application prospects in bone and cartilage tissue engineering. However, further study on the mechanism of related cytokines need to be conducted.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Lang Li
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Ming Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Xin Duan
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Ye Long
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Zhou Xiang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041,
| |
Collapse
|
16
|
D'Este M, Eglin D, Alini M. Lessons to be learned and future directions for intervertebral disc biomaterials. Acta Biomater 2018; 78:13-22. [PMID: 30092378 DOI: 10.1016/j.actbio.2018.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/16/2018] [Accepted: 08/04/2018] [Indexed: 02/07/2023]
Abstract
Biomaterials science has achieved significant advancements for the replacement, repair and regeneration of intervertebral disc tissues. However, the translation of this research to the clinic presents hurdles. The goal of this paper is to identify strategies to recapitulate the intrinsic complexities of the intervertebral disc, to highlight the unresolved issues in basic knowledge hindering the clinical translation, and finally to report on the emerging technologies in the biomaterials field. On this basis, we identify promising research directions, with the hope of stimulating further debate and advances for resolving clinical problems such as cervical and low back pain using biomaterial-based approaches. STATEMENT OF SIGNIFICANCE Although not life-threatening, intervertebral disc disorders have enormous impact on life quality and disability. Disc function within the human body is mainly mechanical, and therefore the use of biomaterials to rescue disc function and alleviate pain is logical. Despite intensive research, the clinical translation of biomaterial-based therapies is hampered by the intrinsic complexity of this organ. After decades of development, artificial discs or tissue replacements are still niche applications given their issues of integration and displacement with detrimental consequences. The struggles of biological therapies and tissue engineering are therefore understandable. However, recent advances in biomaterial science give new hope. In this paper we identify the most promising new directions for intervertebral disc biomaterials.
Collapse
|
17
|
Rigo A, Ferrarini I, Innamorati G, Vinante F. A single amino acid substitution in CXCL12 confers functional selectivity at the beta-arrestin level. Oncotarget 2018; 9:28830-28841. [PMID: 29989007 PMCID: PMC6034749 DOI: 10.18632/oncotarget.25533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/13/2018] [Indexed: 12/31/2022] Open
Abstract
CXCL12/CXCR4 axis relies on both heterotrimeric Gi protein and β-arrestin coupling to trigger downstream responses. G protein activation allows for calcium flux, chemotaxis and early extracellular-signal regulated kinases 1/2 (ERK1/2) phosphorylation, whereas β-arrestin recruitment leads to late signaling, receptor desensitization and internalization. Together they may regulate the balance between transactivation and transinhibition of epithelial growth factor receptor 1 (HER1). Since we have previously noted significant differences between CXCL12 and its structural variant [N33A]CXCL12 in CXCR4 signaling, we sought to better characterize them by performing cAMP inhibition and β-arrestin recruitment assays, as well as functional tests that separately investigate G protein and β-arrestin-induced responses. [N33A]CXCL12 showed reduced potency both in Gαi coupling and β-arrestin recruitment as compared to the wild type chemokine, acting as an unbiased ligand. While these findings translated into reduced potency within Gαi-dependent functions, β-arrestin-dependent modules were affected in a more peculiar way. Unlike CXCL12, the mutant analogue did not restore HB-EGF-stimulated HER1 from CXCR4-induced transinhibition, and did not trigger the late wave of ERK1/2 phosphorylation. Instead, CXCR4 internalization was not impaired upon [N33A]CXCL12 stimulation. These differences highlight the novel opportunity to dissect CXCL12 signaling within the β-arrestin layer, in which the mutant chemokine clearly favors the internalization module over the other pathways. Such functional selectivity has an impact on HER1 activation status and may play a relevant part in the crosstalk between tyrosine kinase and seven transmembrane receptors.
Collapse
Affiliation(s)
- Antonella Rigo
- Section of Hematology, Cancer Research & Cell Biology Laboratory, Department of Medicine, University of Verona, Verona, Italy
| | - Isacco Ferrarini
- Section of Hematology, Cancer Research & Cell Biology Laboratory, Department of Medicine, University of Verona, Verona, Italy
| | - Giulio Innamorati
- Laboratory of Translational Surgery, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Fabrizio Vinante
- Section of Hematology, Cancer Research & Cell Biology Laboratory, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
18
|
Chen G, Fang T, Qi Y, Yin X, Di T, Feng G, Lei Z, Zhang Y, Huang Z. Combined Use of Mesenchymal Stromal Cell Sheet Transplantation and Local Injection of SDF-1 for Bone Repair in a Rat Nonunion Model. Cell Transplant 2018; 25:1801-1817. [PMID: 26883892 DOI: 10.3727/096368916x690980] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone nonunion treatments pose a challenge in orthopedics. This study investigated the joint effects of using mesenchymal stem cell (MSC) sheets with local injection of stromal cell-derived factor-1 (SDF-1) on bone formation. In vitro, we found that migration of MSCs was mediated by SDF-1 in a dose-dependent manner. Moreover, stimulation with SDF-1 had no direct effect on the proliferation or osteogenic differentiation of MSCs. Furthermore, the results indicated elevated expression levels of bone morphogenetic protein 2, alkaline phosphatase, osteocalcin, and vascular endothelial growth factor in MSC sheets compared with MSCs cultured in medium. New bone formation in fractures was evaluated by X-ray, micro-computed tomography (micro-CT), hematoxylin and eosin (H&E) staining, Safranin-O staining, and immunohistochemistry in vivo. In the rat bone fracture model, the MSC sheets transplanted into the injured site along with injection of SDF-1 showed significantly more new bone formation within the gap. Moreover, at 8 weeks, complete bone union was obtained in this group. In contrast, the control group showed nonunion of the bone. Our study suggests a new strategy involving the use of MSC sheets with a local injection of SDF-1 for hard tissue reconstruction, such as the healing of nonunions and bone defects.
Collapse
Affiliation(s)
- Guangnan Chen
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, P.R. China.,Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Tingting Fang
- Liver Cancer Institute, Zhongshan Hospital, Shanghai Medical School of Fudan University, Shanghai, P.R. China
| | - Yiying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Xiaofan Yin
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Tuoyu Di
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Gang Feng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Zhong Lei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Yuxiang Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Zhongming Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China.,Department of Orthopaedic Surgery, Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou, P.R. China.,Department of Orthopaedic Surgery, Xiaoshan Chinese Medical Hospital, Hangzhou, P.R. China.,Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, P.R. China
| |
Collapse
|
19
|
Zhao W, Jin K, Li J, Qiu X, Li S. Delivery of stromal cell-derived factor 1α for in situ tissue regeneration. J Biol Eng 2017; 11:22. [PMID: 28670340 PMCID: PMC5492719 DOI: 10.1186/s13036-017-0058-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/29/2017] [Indexed: 02/06/2023] Open
Abstract
In situ tissue regeneration approach aims to exploit the body's own biological resources and reparative capability and recruit host cells by utilizing cell-instructive biomaterials. In order to immobilize and release bioactive factors in biomaterials, it is important to engineer the load effectiveness, release kinetics and cell recruiting capabilities of bioactive molecules by using suitable bonding strategies. Stromal cell-derived factor 1α (SDF-1α) is one of the most potent chemokines for stem cell recruitment, and SDF-1α-loaded scaffolds have been used for the regeneration of many types of tissues. This review summarizes the strategies to incorporate SDF-1α into scaffolds, including direct loading or adsorption, polyion complexes, specific heparin-mediated interaction and particulate system, which may be applied to the immobilization of other chemokines or growth factors. In addition, we discuss the application of these strategies in the regeneration of tissues such as blood vessel, myocardium, cartilage and bone.
Collapse
Affiliation(s)
- Wen Zhao
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072 China
| | - Kaixiang Jin
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072 China
| | - Jiaojiao Li
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072 China
| | - Xuefeng Qiu
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Song Li
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
20
|
Guo HF, Dai WW, Qian DH, Qin ZX, Lei Y, Hou XY, Wen C. A simply prepared small-diameter artificial blood vessel that promotes in situ endothelialization. Acta Biomater 2017; 54:107-116. [PMID: 28238915 DOI: 10.1016/j.actbio.2017.02.038] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 01/04/2023]
Abstract
Synthetic grafts are of limited use in small-diameter vessels (Φ<6mm) due to the poor patency rate. The inability of such grafts to achieve early endothelialization together with the compliance mismatch between the grafts and the native vessels promote thrombosis, which eventually leads to graft occlusion. In the current study, stromal cell-derived factor (SDF)-1α/vascular endothelial growth factor (VEGF)-loaded polyurethane (PU) conduits were simply prepared via electrospinning. The mechanical property, drug release behavior and cytocompatibility of the conduits were investigated. The effects of the conduits on endothelial progenitor cell (EPC) mobilization and differentiation were examined in vitro. Then, the conduits were implanted as canine femoral artery interposition grafts. The results revealed that SDF-1α and VEGF were quickly released shortly after implantation, and the conduits exhibited slow and sustained release thereafter. The cytokines had definite effects on EPC mobilization and differentiation in vitro and promoted conduit endothelialization in vivo. The conduits had good tissue compatibility and favorable compliance. All of these features inhibited the conduits from being occluded, thereby improving their long-term patency rate. At 6th month postoperatively, 5 of the 8 grafts were patent while all the 8 grafts without the cytokines were occluded. These findings provide a simple and effective method for the construction of small-diameter artificial blood vessels with the aim of facilitating early endothelialization and improving long-term patency. STATEMENT OF SIGNIFICANCE: (1) SDF-1α/VEGF loaded PU conduits were simply prepared by electrospinning. The cytokines with definite and potent effects on angiogenesis were used to avoid complicated mechanism researches. Compared with most of the current vascular grafts which are of poor strength or elasticity, the conduits have favorable mechanical property. All of these inhibit the conduits from occlusion, and thus improve their long-term patency rate. (2) For the in vivo tests, the dogs did not receive any anticoagulant medication in the follow-up period to expose the grafts to the strictest conditions. In vivo endothelialization of the conduits was thoroughly investigated by Sonography, HE staining, SEM and LSCM. The study will be helpful for the construction of small-diameter artificial blood vessels.
Collapse
Affiliation(s)
- Hong-Feng Guo
- Department of General Medicine, Health Service Training Base, Third Military Medical University, Chongqing 400038, China
| | - Wei-Wei Dai
- Department of General Medicine, Health Service Training Base, Third Military Medical University, Chongqing 400038, China
| | - De-Hui Qian
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zhe-Xue Qin
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yan Lei
- Department of General Medicine, Health Service Training Base, Third Military Medical University, Chongqing 400038, China
| | - Xiao-Yu Hou
- Department of General Medicine, Health Service Training Base, Third Military Medical University, Chongqing 400038, China
| | - Can Wen
- Department of General Medicine, Health Service Training Base, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
21
|
Lo SC, Li KC, Chang YH, Hsu MN, Sung LY, Vu TA, Hu YC. Enhanced critical-size calvarial bone healing by ASCs engineered with Cre/loxP-based hybrid baculovirus. Biomaterials 2017; 124:1-11. [DOI: 10.1016/j.biomaterials.2017.01.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/16/2016] [Accepted: 01/27/2017] [Indexed: 12/11/2022]
|
22
|
Mi L, Liu H, Gao Y, Miao H, Ruan J. Injectable nanoparticles/hydrogels composite as sustained release system with stromal cell-derived factor-1α for calvarial bone regeneration. Int J Biol Macromol 2017; 101:341-347. [PMID: 28330754 DOI: 10.1016/j.ijbiomac.2017.03.098] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 01/16/2023]
Abstract
Repair of craniofacial bony defects remains a challenge for surgeons due to the delicate and complex anatomy of the craniofacial skeleton. Stromal cell-derived factor-1α (SDF-1α) is an important chemokine which plays a critical role in the homing of mesenchymal stem cells (MSC), while, the shortcomings including short half-life and easy degradation by enzymes made it in relatively low efficacy. In this work, SDF-1α/chitosan/carboxymeymethy-chitosan nanoparticles (SDF-1α/CS/CMCS NPs) were prepared and characterized for various parameters including morphology, particle size, zeta potential, loading efficiency and the release characteristics from thermosensitive chitosan/β-glycerol phosphate disodium salt (CS/GP) hydrogels. The SDF-1α encapsulated in CS/CMCS NPs within CS/GP hydrogels showed significantly sustained release effect. The cumulative release of SDF-1α was only 40% during 28d. The data from rat calvarial defects model revealed that the SDF-1α/CS/CMCS NPs embedded hydrogels group could significantly promote the new bone formation (38.5±4.5%), compared to that of the SDF-1α embedded hydrogels group (26.3±7.25%, p<0.05) and the control group (8.64±4.8%, p<0.01). Histological data also confirmed this difference. This study demonstrated the potential applications of nanoparticulate injectable hydrogels for sustained release SDF-1α on bone tissue regeneration.
Collapse
Affiliation(s)
- Lei Mi
- Department of Preventive Dentisty, The Research Center of Stomatology, Stomatology Hospital, Xi'an Jiaotong University, No. 98 West-Five Road, Xi'an 710004, China; Department of Stomatology, Yulin First Hospital, No. 93 Yuxi Avenue, Yulin, 719000 Shaanxi Province, China
| | - Huaiqin Liu
- Department of Stomatology, Yulin First Hospital, No. 93 Yuxi Avenue, Yulin, 719000 Shaanxi Province, China
| | - Yu Gao
- Department of Stomatology, Yulin First Hospital, No. 93 Yuxi Avenue, Yulin, 719000 Shaanxi Province, China
| | - Hui Miao
- Department of Periodontology, School of Stomatology, Fourth Military Medical University, No. 145 West Changle Road, Xi'an 710032, China
| | - Jianping Ruan
- Department of Preventive Dentisty, The Research Center of Stomatology, Stomatology Hospital, Xi'an Jiaotong University, No. 98 West-Five Road, Xi'an 710004, China.
| |
Collapse
|
23
|
Shi J, Sun J, Zhang W, Liang H, Shi Q, Li X, Chen Y, Zhuang Y, Dai J. Demineralized Bone Matrix Scaffolds Modified by CBD-SDF-1α Promote Bone Regeneration via Recruiting Endogenous Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:27511-27522. [PMID: 27686136 DOI: 10.1021/acsami.6b08685] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The reconstruction of bone usually depends on substitute transplantation, which has drawbacks including the limited bone substitutes available, comorbidity, immune rejection, and limited endogenous bone regeneration. Here, we constructed a functionalized bone substitute by combining application of the demineralized bone matrix (DBM) and collagen-binding stromal-cell-derived factor-1α (CBD-SDF-1α). DBM was a poriferous and biodegradable bone substitute, derived from bovine bone and consisting mainly of collagen. CBD-SDF-1α could bind to collagen and be controllably released from the DBM to mobilize stem cells. In a rat femur defect model, CBD-SDF-1α-modified DBM scaffolds could efficiently mobilize CD34+ and c-kit+ endogenous stem cells homing to the injured site at 3 days after implantation. According to the data from micro-CT, CBD-SDF-1α-modified DBM scaffolds could help the bone defects rejoin with mineralization accumulated and bone volume expanded. Interestingly, osteoprotegerin (OPG) and osteopontin (OPN) were highly expressed in CBD-SDF-1α group at an early time after implantation, while osteocalcin (OCN) was more expanded. H&E and Masson's trichrome staining showed that the CBD-SDF-1α-modified DBM scaffold group had more osteoblasts and that the bone defect rejoined earlier. The ultimate strength of the regenerated bone was investigated by three-point bending, showing that the CBD-SDF-1α group had superior strength. In conclusion, CBD-SDF-1α-modified DBM scaffolds could promote bone regeneration by recruiting endogenous stem cells.
Collapse
Affiliation(s)
- Jiajia Shi
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China , Hefei 230026, China
| | - Jie Sun
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University , Chongqing 400038, China
| | - Wen Zhang
- Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University , Suzhou 215007, China
| | - Hui Liang
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Qin Shi
- Orthopedic Department, First Affiliated Hospital of Soochow University , Suzhou 215006, China
| | - Xiaoran Li
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Suzhou Key Laboratory for Nanotheranostics, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University , Chongqing 400038, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|
24
|
Wang T, He J, Zhang Y, Shi W, Dong J, Pei M, Zhu L. A Selective Cell Population from Dermis Strengthens Bone Regeneration. Stem Cells Transl Med 2016; 6:306-315. [PMID: 28170187 PMCID: PMC5442747 DOI: 10.5966/sctm.2015-0426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/22/2016] [Indexed: 11/16/2022] Open
Abstract
Finding appropriate seed cells for bone tissue engineering remains a significant challenge. Considering that skin is the largest organ, we hypothesized that human bone morphogenetic protein receptor type IB (BmprIB)+ dermal cells could have enhanced osteogenic capacity in the healing of critical-sized calvarial defects in an immunodeficient mouse model. In this study, immunohistochemical staining revealed that BmprIB was expressed throughout reticular dermal cells; the positive expression rate of BmprIB was 3.5% ± 0.4% in freshly separated dermal cells, by flow cytometry. Furthermore, in vitro osteogenic capacity of BmprIB+ cells was confirmed by osteogenic-related staining and marker gene expression compared with unsorted dermal cells. In vivo osteogenic capacity was demonstrated by implantation of human BmprIB+ cell/coral constructs in the treatment of 4-mm diameter calvarial defects in an immunodeficient mouse model compared with implantation of unsorted cell/coral constructs and coral scaffold alone. These results indicate that the selective cell population BmprIB from human dermis is a promising osteogenic progenitor cell that can be a large-quantity and high-quality cell source for bone tissue engineering and regeneration. Stem Cells Translational Medicine 2017;6:306-315.
Collapse
Affiliation(s)
- Tingliang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jinguang He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiasheng Dong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, and Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia, USA
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
25
|
Mele L, Vitiello PP, Tirino V, Paino F, De Rosa A, Liccardo D, Papaccio G, Desiderio V. Changing Paradigms in Cranio-Facial Regeneration: Current and New Strategies for the Activation of Endogenous Stem Cells. Front Physiol 2016; 7:62. [PMID: 26941656 PMCID: PMC4764712 DOI: 10.3389/fphys.2016.00062] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022] Open
Abstract
Craniofacial area represent a unique district of human body characterized by a very high complexity of tissues, innervation and vascularization, and being deputed to many fundamental function such as eating, speech, expression of emotions, delivery of sensations such as taste, sight, and earing. For this reasons, tissue loss in this area following trauma or for example oncologic resection, have a tremendous impact on patients' quality of life. In the last 20 years regenerative medicine has emerged as one of the most promising approach to solve problem related to trauma, tissue loss, organ failure etc. One of the most powerful tools to be used for tissue regeneration is represented by stem cells, which have been successfully implanted in different tissue/organs with exciting results. Nevertheless, both autologous and allogeneic stem cell transplantation raise many practical and ethical concerns that make this approach very difficult to apply in clinical practice. For this reason different cell free approaches have been developed aiming to the mobilization, recruitment, and activation of endogenous stem cells into the injury site avoiding exogenous cells implant but instead stimulating patients' own stem cells to repair the lesion. To this aim many strategies have been used including functionalized bioscaffold, controlled release of stem cell chemoattractants, growth factors, BMPs, Platelet-Rich-Plasma, and other new strategies such as ultrasound wave and laser are just being proposed. Here we review all the current and new strategies used for activation and mobilization of endogenous stem cells in the regeneration of craniofacial tissue.
Collapse
Affiliation(s)
- Luigi Mele
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Pietro Paolo Vitiello
- Medical Oncology, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara," Second University of Naples Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Francesca Paino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Alfredo De Rosa
- Department of Odontology and Surgery, Second University of Naples Naples, Italy
| | - Davide Liccardo
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| |
Collapse
|
26
|
Chen G, Yang L, Lv Y. Cell-free scaffolds with different stiffness but same microstructure promote bone regeneration in rabbit large bone defect model. J Biomed Mater Res A 2015; 104:833-41. [PMID: 26650620 DOI: 10.1002/jbm.a.35622] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/31/2015] [Accepted: 12/02/2015] [Indexed: 12/26/2022]
Abstract
To promote bone healing, bone repair biomaterials are increasingly designed to incorporate growth factors. However, the impact of matrix mechanics of cell-free scaffold independent of microstructure on the osteogenic differentiation of endogenous osteoprogenitor cells orchestrating bone repair and regeneration remains not to be fully understood. In our recent study, three-dimensional (3D) scaffolds with different stiffness but same microstructure have been successfully fabricated by coating decellularized bone with collagen/hydroxyapatite (HA) mixture with different collagen rations. It has been demonstrated that the scaffold with optimal stiffness can induce the osteogenic differentiation of MSCs in vitro and in the subcutaneous tissue. The present in vivo study further investigated the repair efficiency of these scaffolds in a rabbit radius with a critical-sized segmental defect model and its potential mechanism. Micro-computed tomography (μ-CT), X-ray and histological analysis were carried out to evaluate the repair capacity of these scaffolds. The results demonstrated that the cell-free scaffold with optimal stiffness incorporation of endogenous osteoprogenitor cells significantly promoted the repair and reconstruction quality of mass bone defect. One of the crucial mechanisms was that hypoxia and stromal cell-derived factor-1α (SDF-1α) mediated mesenchymal stem cells (MSCs) migration by which matrix mechanics exerted influence on bone fracture healing. These findings suggested that only modulating the matrix stiffness of cell-free scaffold can be one of the most attractive strategies for promoting the progression of bone healing.
Collapse
Affiliation(s)
- Guobao Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| |
Collapse
|
27
|
Yasui T, Mabuchi Y, Toriumi H, Ebine T, Niibe K, Houlihan DD, Morikawa S, Onizawa K, Kawana H, Akazawa C, Suzuki N, Nakagawa T, Okano H, Matsuzaki Y. Purified Human Dental Pulp Stem Cells Promote Osteogenic Regeneration. J Dent Res 2015; 95:206-14. [PMID: 26494655 DOI: 10.1177/0022034515610748] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human dental pulp stem/progenitor cells (hDPSCs) are attractive candidates for regenerative therapy because they can be easily expanded to generate colony-forming unit-fibroblasts (CFU-Fs) on plastic and the large cell numbers required for transplantation. However, isolation based on adherence to plastic inevitably changes the surface marker expression and biological properties of the cells. Consequently, little is currently known about the original phenotypes of tissue precursor cells that give rise to plastic-adherent CFU-Fs. To better understand the in vivo functions and translational therapeutic potential of hDPSCs and other stem cells, selective cell markers must be identified in the progenitor cells. Here, we identified a dental pulp tissue-specific cell population based on the expression profiles of 2 cell-surface markers LNGFR (CD271) and THY-1 (CD90). Prospectively isolated, dental pulp-derived LNGFR(Low+)THY-1(High+) cells represent a highly enriched population of clonogenic cells--notably, the isolated cells exhibited long-term proliferation and multilineage differentiation potential in vitro. The cells also expressed known mesenchymal cell markers and promoted new bone formation to heal critical-size calvarial defects in vivo. These findings suggest that LNGFR(Low+)THY-1(High+) dental pulp-derived cells provide an excellent source of material for bone regenerative strategies.
Collapse
Affiliation(s)
- T Yasui
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan Department of Dentistry and Oral Surgery, Kawasaki Municipal Kawasaki Hospital, Kanagawa, Japan
| | - Y Mabuchi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Toriumi
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - T Ebine
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - K Niibe
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan Division of Molecular and Regenerative Prosthodontics, Graduate School of Dentistry, Tohoku University, Miyagi, Japan
| | - D D Houlihan
- Centre for Liver Research, NIHR Biomedical Research Unit, University of Birmingham, Birmingham, UK
| | - S Morikawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | - K Onizawa
- Department of Dentistry and Oral Surgery, Kawasaki Municipal Kawasaki Hospital, Kanagawa, Japan
| | - H Kawana
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | - C Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - N Suzuki
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - T Nakagawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, Tokyo, Japan
| | - H Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Y Matsuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan Department of Cancer Biology, Faculty of Medicine, Shimane University, Shimane, Japan
| |
Collapse
|
28
|
Hwang HD, Lee JT, Koh JT, Jung HM, Lee HJ, Kwon TG. Sequential Treatment with SDF-1 and BMP-2 Potentiates Bone Formation in Calvarial Defects. Tissue Eng Part A 2015; 21:2125-35. [PMID: 25919507 DOI: 10.1089/ten.tea.2014.0571] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1) protein and its receptor, CXCR-4, play an important role in tissue repair and regeneration in various organs, including the bone. SDF-1 is indispensable for bone morphogenetic protein-2 (BMP-2)-induced osteogenic differentiation. However, SDF-1 is not needed after the osteogenic induction has been activated. Since the precise condition for the additive effects of combined DF-1 and BMP-2 in bone healing had not been fully investigated, we aimed to determine the optimal conditions for SDF-1- and BMP-2-mediated bone regeneration. We examined the in vitro osteoblastic differentiation and cell migration after sequential treatments with SDF-1 and BMP-2. Based on the in vitro additive effects of SDF-1 and BMP-2, the critical size defects of mice calvaria were treated with these cytokines in various sequences. Phosphate buffered saline (PBS)-, SDF-1-, or BMP-2-soaked collagen scaffolds were implanted into the calvarial defects (n=36). Periodic percutaneous injections of PBS or the cytokine SDF-1 and BMP-2 into the implanted scaffolds were performed on days 3 and 6, postoperatively. Six experimental groups were used according to the types and sequences of the cytokine treatments. After 28 days, the mice were euthanized and bone formation was evaluated with microcomputed tomography and histology. The molecular mechanism of the additive effect of SDF-1 and BMP-2 was evaluated by analyzing intracellular signal transduction through Smad and Erk phosphorylation. The in vitro experiments revealed that, among all the treatments, the treatment with BMP-2 after SDF-1 showed the strongest osteoblastic differentiation and enhanced cell migration. Similarly, in the animal model, the treatment with SDF-1 followed by BMP-2 treatment showed the highest degree of new bone regeneration than any other groups, including the one with continuous BMP-2 treatment. This new bone formation can be partially explained by the activation of Smad and Erk pathways and enhanced cell migration. These results suggest that sequential treatment with the cytokines, SDF-1 and BMP-2, may be a promising strategy for accelerating bone regeneration in critical size defects.
Collapse
Affiliation(s)
- Hee-Don Hwang
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University , Daegu, Republic of Korea
| | - Jung-Tae Lee
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University , Daegu, Republic of Korea
| | - Jeong-Tae Koh
- 2 Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University , Gwangju, Republic of Korea
| | - Hong-Moon Jung
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University , Daegu, Republic of Korea
| | - Heon-Jin Lee
- 3 Department of Oral Microbiology, School of Dentistry, Kyungpook National University , Daegu, Republic of Korea
| | - Tae-Geon Kwon
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University , Daegu, Republic of Korea
| |
Collapse
|
29
|
Herrmann M, Verrier S, Alini M. Strategies to Stimulate Mobilization and Homing of Endogenous Stem and Progenitor Cells for Bone Tissue Repair. Front Bioeng Biotechnol 2015; 3:79. [PMID: 26082926 PMCID: PMC4451737 DOI: 10.3389/fbioe.2015.00079] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/16/2015] [Indexed: 12/17/2022] Open
Abstract
The gold standard for the treatment of critical-size bone defects is autologous or allogenic bone graft. This has several limitations including donor site morbidity and the restricted supply of graft material. Cell-based tissue engineering strategies represent an alternative approach. Mesenchymal stem cells (MSCs) have been considered as a source of osteoprogenitor cells. More recently, focus has been placed on the use of endothelial progenitor cells (EPCs), since vascularization is a critical step in bone healing. Although many of these approaches have demonstrated effectiveness for bone regeneration, cell-based therapies require time consuming and cost-expensive in vitro cell expansion procedures. Accordingly, research is becoming increasingly focused on the homing and stimulation of native cells. The stromal cell-derived factor-1 (SDF-1) - CXCR4 axis has been shown to be critical for the recruitment of MSCs and EPCs. Vascular endothelial growth factor (VEGF) is a key factor in angiogenesis and has been targeted in many studies. Here, we present an overview of the different approaches for delivering homing factors to the defect site by absorption or incorporation to biomaterials, gene therapy, or via genetically manipulated cells. We further review strategies focusing on the stimulation of endogenous cells to support bone repair. Finally, we discuss the major challenges in the treatment of critical-size bone defects and fracture non-unions.
Collapse
Affiliation(s)
| | | | - Mauro Alini
- AO Research Institute Davos , Davos , Switzerland
| |
Collapse
|
30
|
Effect of dual treatment with SDF-1 and BMP-2 on ectopic and orthotopic bone formation. PLoS One 2015; 10:e0120051. [PMID: 25781922 PMCID: PMC4363323 DOI: 10.1371/journal.pone.0120051] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/02/2015] [Indexed: 01/07/2023] Open
Abstract
Purposes The potent stem cell homing factor stromal cell-derived factor-1 (SDF-1) actively recruits mesenchymal stem cells from circulation and from local bone marrow. It is well established that bone morphogenetic protein-2 (BMP-2) induces ectopic and orthotopic bone formation. However, the exact synergistic effects of BMP-2 and SDF-1 in ectopic and orthotopic bone regeneration models have not been fully investigated. The purpose of this study was to evaluate the potential effects of simultaneous SDF-1 and BMP-2 treatment on bone formation. Materials and Methods Various doses of SDF-1 were loaded onto collagen sponges with or without BMP-2.These sponges were implanted into subcutaneous pockets and critical-size calvarial defects in C57BL/6 mice. The specimens were harvested 4 weeks post-surgery and the degree of bone formation in specimens was evaluated by histomorphometric and radiographic density analyses. Osteogenic potential and migration capacity of mesenchymal cells and capillary tube formation of endothelial cells following dual treatment with SDF-1 and BMP-2 were evaluated with in vitro assays. Results SDF-1-only-treated implants did not yield significant in vivo bone formation and SDF-1 treatment did not enhance BMP-2-induced ectopic and orthotopic bone regeneration. In vitro experiments showed that concomitant use of BMP-2 and SDF-1 had no additive effect on osteoblastic differentiation, cell migration or angiogenesis compared to BMP-2 or SDF-1 treatment alone. Conclusions These findings imply that sequence-controlled application of SDF-1 and BMP-2 must be further investigated for the enhancement of robust osteogenesis in bone defects.
Collapse
|
31
|
Kyllönen L, D’Este M, Alini M, Eglin D. Local drug delivery for enhancing fracture healing in osteoporotic bone. Acta Biomater 2015; 11:412-34. [PMID: 25218339 DOI: 10.1016/j.actbio.2014.09.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/30/2014] [Accepted: 09/04/2014] [Indexed: 01/08/2023]
Abstract
Fragility fractures can cause significant morbidity and mortality in patients with osteoporosis and inflict a considerable medical and socioeconomic burden. Moreover, treatment of an osteoporotic fracture is challenging due to the decreased strength of the surrounding bone and suboptimal healing capacity, predisposing both to fixation failure and non-union. Whereas a systemic osteoporosis treatment acts slowly, local release of osteogenic agents in osteoporotic fracture would act rapidly to increase bone strength and quality, as well as to reduce the bone healing period and prevent development of a problematic non-union. The identification of agents with potential to stimulate bone formation and improve implant fixation strength in osteoporotic bone has raised hope for the fast augmentation of osteoporotic fractures. Stimulation of bone formation by local delivery of growth factors is an approach already in clinical use for the treatment of non-unions, and could be utilized for osteoporotic fractures as well. Small molecules have also gained ground as stable and inexpensive compounds to enhance bone formation and tackle osteoporosis. The aim of this paper is to present the state of the art on local drug delivery in osteoporotic fractures. Advantages, disadvantages and underlying molecular mechanisms of different active species for local bone healing in osteoporotic bone are discussed. This review also identifies promising new candidate molecules and innovative approaches for the local drug delivery in osteoporotic bone.
Collapse
|