1
|
Shariati K, Bedar M, Huang KX, Moghadam S, Mirzaie S, LaGuardia JS, Chen W, Kang Y, Ren X, Lee JC. Biomaterial Cues for Regulation of Osteoclast Differentiation and Function in Bone Regeneration. ADVANCED THERAPEUTICS 2025; 8:2400296. [PMID: 39867107 PMCID: PMC11756815 DOI: 10.1002/adtp.202400296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 01/28/2025]
Abstract
Tissue regeneration involves dynamic dialogue between and among different cells and their surrounding matrices. Bone regeneration is specifically governed by reciprocity between osteoblasts and osteoclasts within the bone microenvironment. Osteoclast-directed resorption and osteoblast-directed formation of bone are essential to bone remodeling, and the crosstalk between these cells is vital to curating a sequence of events that culminate in the creation of bone tissue. Among bone biomaterial strategies, many have investigated the use of different material cues to direct the development and activity of osteoblasts. However, less attention has been given to exploring features that similarly target osteoclast formation and activity, with even fewer strategies demonstrating or integrating biomaterial-directed modulation of osteoblast-osteoclast coupling. This review aims to describe various biomaterial cues demonstrated to influence osteoclastogenesis and osteoclast function, emphasizing those that enhance a material construct's ability to achieve bone healing and regeneration. Additionally discussed are approaches that influence the communication between osteoclasts and osteoblasts, particularly in a manner that takes advantage of their coupling. Deepening our understanding of how biomaterial cues may dictate osteoclast differentiation, function, and influence on the microenvironment may enable the realization of bone-replacement interventions with enhanced integrative and regenerative capacities.
Collapse
Affiliation(s)
- Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Sarah Mirzaie
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
2
|
Chan ME, Ashdown C, Strait L, Pasumarthy S, Hassan A, Crimarco S, Singh C, Patel VS, Pagnotti G, Khan O, Uzer G, Rubin CT. Low intensity mechanical signals promote proliferation in a cell-specific manner: Tailoring a non-drug strategy to enhance biomanufacturing yields. MECHANOBIOLOGY IN MEDICINE 2024; 2:100080. [PMID: 39717386 PMCID: PMC11666124 DOI: 10.1016/j.mbm.2024.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Biomanufacturing relies on living cells to produce biotechnology-based therapeutics, tissue engineering constructs, vaccines, and a vast range of agricultural and industrial products. With the escalating demand for these bio-based products, any process that could improve yields and shorten outcome timelines by accelerating cell proliferation would have a significant impact across the discipline. While these goals are primarily achieved using biological or chemical strategies, harnessing cell mechanosensitivity represents a promising - albeit less studied - physical pathway to promote bioprocessing endpoints, yet identifying which mechanical parameters influence cell activities has remained elusive. We tested the hypothesis that mechanical signals, delivered non-invasively using low-intensity vibration (LIV; <1 g, 10-500 Hz), will enhance cell expansion, and determined that any unique signal configuration was not equally influential across a range of cell types. Varying frequency, intensity, duration, refractory period, and daily doses of LIV increased proliferation in Chinese Hamster Ovary (CHO)-adherent cells (+79% in 96 hr) using a particular set of LIV parameters (0.2 g, 500 Hz, 3 × 30 min/d, 2 hr refractory period), yet this same mechanical input suppressed proliferation in CHO-suspension cells (-13%). Another set of LIV parameters (30 Hz, 0.7 g, 2 × 60 min/d, 2 hr refractory period) however, were able to increase the proliferation of CHO-suspension cells by 210% and T-cells by 20.3%. Importantly, we also reported that T-cell response to LIV was in-part dependent upon AKT phosphorylation, as inhibiting AKT phosphorylation reduced the proliferative effect of LIV by over 60%, suggesting that suspension cells utilize mechanism(s) similar to adherent cells to sense specific LIV signals. Particle image velocimetry combined with finite element modeling showed high transmissibility of these signals across fluids (>90%), and LIV effectively scaled up to T75 flasks. Ultimately, when LIV is tailored to the target cell population, it's highly efficient transmission across media represents a means to non-invasively augment biomanufacturing endpoints for both adherent and suspended cells, and holds immediate applications, ranging from small-scale, patient-specific personalized medicine to large-scale commercial biocentric production challenges.
Collapse
Affiliation(s)
- M. Ete Chan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Christopher Ashdown
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
- Medical Scientist Training Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Lia Strait
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Sishir Pasumarthy
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Abdullah Hassan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Steven Crimarco
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Chanpreet Singh
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Vihitaben S. Patel
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Gabriel Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Omor Khan
- Department of Mechanical and Biomedical Engineering, College of Engineering, Boise State University, Boise, ID, 83725-205, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, College of Engineering, Boise State University, Boise, ID, 83725-205, USA
| | - Clinton T. Rubin
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
- Center for Biotechnology, New York State Center for Advanced Technology in Medical Biotechnology, Stony Brook University, Stony Brook, NY, 11794-5281, USA
| |
Collapse
|
3
|
He S, Hu R, Yao X, Cui J, Liu H, Zhu M, Ning L. The effects of heat and hydrogen peroxide treatment on the osteoinductivity of demineralized cortical bone: a potential method for preparing tendon/ligament repair scaffolds. Regen Biomater 2024; 11:rbae116. [PMID: 39398284 PMCID: PMC11471265 DOI: 10.1093/rb/rbae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/07/2024] [Indexed: 10/15/2024] Open
Abstract
Recent studies have indicated that demineralized cortical bone (DCB) may be used to repair tendons and ligaments, such as the patellar tendon and anterior cruciate ligament (ACL). Hydrogen peroxide (H2O2) has been shown to reduce the osteoinductivity of DCB, and heat treatment may also decrease the osteoinductivity of DCB. The purpose of this study was (i) to determine whether heat treatment reduces the osteoinductivity of DCB and (ii) to compare the effectiveness of heat treatment and H2O2 treatment on BMP-2 inactivation. DCB was prepared by immersion in 0.6 N hydrochloric acid, and DCB-H and DCB-HO were prepared by heat treatment (70°C for 8 h) and H2O2 treatment (3% H2O2 for 8 h), respectively. The surface topographies, elemental distributions and histological structures of the scaffolds were observed by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FT-IR) and histological staining. The viability and osteogenic differentiation of TDSCs cultured on the scaffolds were evaluated via live/dead cell staining and Cell Counting Kit-8 (CCK-8) testing, real-time polymerase chain reaction (RT-PCR) and western bolt (WB) analysis, alkaline phosphatase activity (ALP) and alizarin red S (ARS) staining. The intramuscular implantation of the scaffolds in rats was also used to evaluate the effect of heat treatment and H2O2 treatment on the osteoinductivity of DCB. Our results demonstrated that both treatments removed BMP-2 and osteocalcin (OCN) within the DCB and that DCB-H and DCB-HO had good cytocompatibility and reduced the osteogenic differentiation of TDSCs. Moreover, the in vivo results indicated that the DCB-H and DCB-HO groups had smaller areas of osteoid formation than did the DCB group, and the DCB-HO group had the smallest area among the three groups. Our study demonstrated that heat treatment could reduce the osteoinductivity of DCB, and that H2O2 treatment was more effective than heat treatment.
Collapse
Affiliation(s)
- Shukun He
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ruonan Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuan Yao
- Department of Clinical Hematology, Faculty of Laboratory Medicine, Army Medical University, Chongqing, 400038, China
| | - Jing Cui
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huimin Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Zhu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangju Ning
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Chittoria A, Malviya Y, Adurti A. Sinus Savvy: Exploring the Current Techniques of Maxillary Sinus Augmentation. Cureus 2024; 16:e61933. [PMID: 38978885 PMCID: PMC11230615 DOI: 10.7759/cureus.61933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2024] [Indexed: 07/10/2024] Open
Abstract
Sinus ridge augmentation is a surgical procedure aimed at increasing the volume of bone in the posterior maxilla to permit successful dental implant placement. The current review article presents an overview of various techniques used for sinus ridge augmentation, including the lateral window technique, crestal approach, transalveolar technique, and piezoelectric osteotomy. The article examines the advantages and limitations of each technique, such as invasiveness, surgical difficulty, and the requirement for additional procedures. Additionally, the article discusses the factors that influence the success of the procedure, including patient age, residual bone height, and the kind of bone graft substance used. The review also emphasizes the importance of proper case selection, surgical planning, and postoperative care to ensure optimal outcomes. Overall, the article provides valuable insights into the current techniques used for sinus ridge augmentation, highlighting the need for further research to improve patient outcomes and the success of placing dental implants over the long run.
Collapse
Affiliation(s)
| | - Yogendra Malviya
- Oral and Maxillofacial Surgery, Jaipur Dental College, Jaipur, IND
| | - Aparajita Adurti
- Oral and Maxillofacial Surgery, School of Dental Sciences, Sharda University, Noida, IND
| |
Collapse
|
5
|
Li X, Zhang C, Feng C, Zhang Z, Feng N, Sha H, Luo X, Zou G, Liang H. Transcriptome Analysis Elucidates the Potential Key Genes Involved in Rib Development in bmp6-Deficient Silver Carp ( Hypophthalmichthys molitrix). Animals (Basel) 2024; 14:1451. [PMID: 38791669 PMCID: PMC11117292 DOI: 10.3390/ani14101451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Bone morphogenetic protein 6 (BMP-6) is a constituent of the TGF-β superfamily, known for its ability to stimulate bone and cartilage formation. The investigation of bmp6's involvement in the formation of intermuscular bones in fish has garnered significant attention in recent years. The rib cage is an important skeletal structure that plays a protective function for internal organs in fish. However, there has been limited research conducted on the effects of the bmp6 gene on rib development. Silver carp is one of four major fish in China, favoured for its affordability and tender muscle. Nevertheless, the presence of numerous intermuscular bones in silver carp significantly hinders the advancement of its palatability and suitability for processing. This study showcases the effective utilisation of CRISPR/Cas9 technology for the purpose of disrupting the bmp6 gene in silver carp, leading to the creation of chimeras in the P0 generation, marking the first instance of such an achievement. The chimeras exhibited complete viability, normal appearance, and partial intermuscular bones loss, with approximately 30% of them displaying rib bifurcation or bending. Subsequently, a transcriptome analysis on ribs of P0 chimeras and wild-type silver carp was conducted, leading to the identification of 934 genes exhibiting differential expression, of which 483 were found to be up-regulated and 451 were found to be down-regulated. The results of the KEGG analysis revealed that the "NF-kappa B signalling pathway", "Hippo signalling pathway", "osteoclast differentiation", and "haematopoietic cell lineage" exhibited enrichment and displayed a significant correlation with bone development. The up-regulated genes such as tnfα, fos, and ctgf in pathways may facilitate the proliferation and differentiation of osteoclasts, whereas the down-regulation of genes such as tgfb2 and tgfbr1 in pathways may hinder the formation and specialisation of osteoblasts, ultimately resulting in rib abnormalities. This study presents novel findings on the impact of bmp6 gene deletion on the rib development of silver carp, while simultaneously investigating the previously unexplored molecular mechanisms underlying rib defects in fish.
Collapse
Affiliation(s)
- Xiaohui Li
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Chunyan Zhang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Cui Feng
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Zewen Zhang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Nannan Feng
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Laboratory of Zooligical Systematics and Application of Hebei Province, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Hang Sha
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Xiangzhong Luo
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Guiwei Zou
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
| | - Hongwei Liang
- Yangtze River Fisheries Research Institude, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (X.L.); (C.Z.); (C.F.); (Z.Z.); (N.F.); (H.S.); (X.L.); (G.Z.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
6
|
Prodoehl JA, Bakaes Y, Tucker M, Voss F. Off-label: The results of adjunctive bone morphogenetic protein for challenging femur fractures; a review of two cases. Trauma Case Rep 2024; 50:100979. [PMID: 38357291 PMCID: PMC10863425 DOI: 10.1016/j.tcr.2024.100979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
Background Although bone morphogenetic proteins (BMPs) are used as an adjunct to promote healing, they may have unintended effects such as heterotopic ossification (HO). The literature is limited regarding the effect of using off-label BMPs for femur fractures. Case presentation We report two outcomes after off-label use of BMPs for the treatment of femur fractures and propose a possible explanation for the difference. Conclusions BMPs are critical osteoinductive factors in injured bone and muscle that facilitate bony healing. However, it may be important to recognize the potentially negative effects of adding BMP to bone graft material in certain cases to stimulate bone repair. We hope this case series helps surgeons consider the risks and benefits of using BMP for femur fractures, and therefore to decide with caution when BMP is indicated.
Collapse
Affiliation(s)
- Julia Anne Prodoehl
- Prisma Health Midlands Department of Orthopaedic Surgery, 2 Medical Park Rd, Columbia, SC 29203, United States of America
| | - Yianni Bakaes
- University of South Carolina School of Medicine Columbia, 2 Medical Park Rd, Columbia, SC 29203, United States of America
| | - Michael Tucker
- Prisma Health Midlands Department of Orthopaedic Surgery, 2 Medical Park Rd, Columbia, SC 29203, United States of America
| | - Frank Voss
- Medical University of South Carolina Department of Orthopaedic Surgery, 135 Rutledge Ave., Charleston, SC 29425, United States of America
| |
Collapse
|
7
|
Ripamonti U, Duarte R. Mechanistic insights into the spontaneous induction of bone formation. BIOMATERIALS ADVANCES 2024; 158:213795. [PMID: 38335762 DOI: 10.1016/j.bioadv.2024.213795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
The grand discovery of morphogens, or "form-generating substances", revealed that tissue morphogenesis is initiated by soluble molecular signals or morphogens primarily belonging to the transforming growth factor-β (TGF-β) supergene family. The regenerative potential of bone rests on its extracellular matrix, which is the repository of several morphogens that tightly control cellular differentiating pathways, cellular matrix deposition and remodeling. Alluringly, the matrix also contains specific factors transferred from the heterotopic implanted bone matrices initiating "Tissue Induction", as provocatively described in Nature in 1945. Later, it was found that selected genes and gene products of the TGF-β supergene family singly, synchronously, and synergistically mastermind the induction of bone formation. This review describes the phenomenon of the spontaneous and/or intrinsic osteoinductivity of calcium phosphate-based biomaterials and titanium' constructs without the applications of soluble osteogenetic molecular signals. The review shows the spontaneous induction of bone formation initiated by Ca++ activating stem cell differentiation and up-regulation of bone morphogenetic proteins genes. Expressed gene products are embedded into the concavities of the calcium phosphate-based substrata, initiating bone formation as a secondary response. Pure titanium's substrata do not initiate the spontaneous induction of bone formation. The induction of bone is solely dependent on acid, alkali and heat treatments to form apatite layers on the treated titanium surfaces. The induction of bone formation is achieved exclusively by apatite-based biomaterial surfaces. The hydroxyapatite, in its various forms and geometric configurations, finely tunes the induction of bone formation in heterotopic sites. Cellular differentiation by fine-tuning of the cellular molecular machinery is initiated by specific geometric modularity of the hydroxyapatite substrata that push cellular buttons that start the ripple-like cascade of "Tissue Induction", generating newly formed ossicles with bone marrow in heterotopic extraskeletal sites. The highlighted mechanistic insights into the spontaneous induction of bone formation are a research platform invocating selected molecular elements to construct the induction of bone formation.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Raquel Duarte
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Internal Medicine Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
8
|
Szilágyi SS, Burdzinski W, Jatzlau J, Ehrlich M, Knaus P, Henis YI. The Activation of the Fibrodysplasia Ossificans Progressiva-Inducing ALK2-R206H Mutant Depends on the Distinct Homo-Oligomerization Patterns of ACVR2B and ACVR2A. Cells 2024; 13:221. [PMID: 38334613 PMCID: PMC10854824 DOI: 10.3390/cells13030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Mutations in activin-like kinase 2 (ALK2), e.g., ALK2-R206H, induce aberrant signaling to SMAD1/5/8, leading to Fibrodysplasia Ossificans Progressiva (FOP). In spite of extensive studies, the underlying mechanism is still unclear. Here, we quantified the homomeric and heteromeric interactions of ACVR2A, ACVR2B, ALK2-WT, and ALK2-R206H by combining IgG-mediated immobilization of one receptor with fluorescence recovery after photobleaching (FRAP) measurements on the lateral diffusion of a co-expressed receptor. ACVR2B formed stable homomeric complexes that were enhanced by Activin A (ActA), while ACVR2A required ActA for homodimerization. ALK2-WT, but not ALK2-R206H, exhibited homomeric complexes unaffected by ActA. ACVR2B formed ActA-enhanced heterocomplexes with ALK2-R206H or ALK2-WT, while ACVR2A interacted mainly with ALK2-WT. The extent of the homomeric complex formation of ACVR2A or ACVR2B was reflected in their ability to induce the oligomerization of ALK2-R206H and ALK2-WT. Thus, ACVR2B, which forms dimers without ligand, induced ActA-independent ALK2-R206H clustering but required ActA for enhancing the oligomerization of the largely dimeric ALK2-WT. In contrast, ACVR2A, which undergoes homodimerization in response to ActA, required ActA to induce ALK2-R206H oligomerization. To investigate whether these interactions are translated into signaling, we studied signaling by the FOP-inducing hyperactive ALK2-R206H mutant, with ALK2-WT signaling as control. The activation of SMAD1/5/8 signaling in cells expressing ALK2-R206H alone or together with ACVR2A or ACVR2B was measured by blotting for pSMAD1/5/8 and by transcriptional activation assays using BRE-Luc reporter. In line with the biophysical studies, ACVR2B activated ALK2-R206H without ligand, while activation by ACVR2A was weaker and required ActA. We propose that the homodimerization of ACVR2B or ACVR2A dictates their ability to recruit ALK2-R206H into higher complexes, enabling the homomeric interactions of ALK2-R206H receptors and, subsequently, their activation.
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Wiktor Burdzinski
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
9
|
Manole CG, Soare C, Ceafalan LC, Voiculescu VM. Platelet-Rich Plasma in Dermatology: New Insights on the Cellular Mechanism of Skin Repair and Regeneration. Life (Basel) 2023; 14:40. [PMID: 38255655 PMCID: PMC10817627 DOI: 10.3390/life14010040] [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: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
The skin's recognised functions may undergo physiological alterations due to ageing, manifesting as varying degrees of facial wrinkles, diminished tautness, density, and volume. Additionally, these functions can be disrupted (patho)physiologically through various physical and chemical injuries, including surgical trauma, accidents, or chronic conditions like ulcers associated with diabetes mellitus, venous insufficiency, or obesity. Advancements in therapeutic interventions that boost the skin's innate regenerative abilities could significantly enhance patient care protocols. The application of Platelet-Rich Plasma (PRP) is widely recognized for its aesthetic and functional benefits to the skin. Yet, the endorsement of PRP's advantages often borders on the dogmatic, with its efficacy commonly ascribed solely to the activation of fibroblasts by the factors contained within platelet granules. PRP therapy is a cornerstone of regenerative medicine which involves the autologous delivery of conditioned plasma enriched by platelets. This is achieved by centrifugation, removing erythrocytes while retaining platelets and their granules. Despite its widespread use, the precise sequences of cellular activation, the specific cellular players, and the molecular machinery that drive PRP-facilitated healing are still enigmatic. There is still a paucity of definitive and robust studies elucidating these mechanisms. In recent years, telocytes (TCs)-a unique dermal cell population-have shown promising potential for tissue regeneration in various organs, including the dermis. TCs' participation in neo-angiogenesis, akin to that attributed to PRP, and their role in tissue remodelling and repair processes within the interstitia of several organs (including the dermis), offer intriguing insights. Their potential to contribute to, or possibly orchestrate, the skin regeneration process following PRP treatment has elicited considerable interest. Therefore, pursuing a comprehensive understanding of the cellular and molecular mechanisms at work, particularly those involving TCs, their temporal involvement in structural recovery following injury, and the interconnected biological events in skin wound healing and regeneration represents a compelling field of study.
Collapse
Affiliation(s)
- Catalin G. Manole
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M. Voiculescu
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
10
|
Jaber M, Hofbauer LC, Hofbauer C, Duda GN, Checa S. Reduced Bone Regeneration in Rats With Type 2 Diabetes Mellitus as a Result of Impaired Stromal Cell and Osteoblast Function-A Computer Modeling Study. JBMR Plus 2023; 7:e10809. [PMID: 38025037 PMCID: PMC10652174 DOI: 10.1002/jbm4.10809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 12/01/2023] Open
Abstract
Bone has the fascinating ability to self-regenerate. However, under certain conditions, such as type 2 diabetes mellitus (T2DM), this ability is impaired. T2DM is a chronic metabolic disease known by the presence of elevated blood glucose levels that is associated with reduced bone regeneration capability, high fracture risk, and eventual non-union risk after a fracture. Several mechanical and biological factors relevant to bone regeneration have been shown to be affected in a diabetic environment. However, whether impaired bone regeneration in T2DM can be explained due to mechanical or biological alterations remains unknown. To elucidate the relevance of either one, the aim of this study was to investigate the relative contribution of T2DM-related alterations on either cellular activity or mechanical stimuli driving bone regeneration. A previously validated in silico computer modeling approach that was capable of explaining bone regeneration in uneventful conditions of healing was further developed to investigate bone regeneration in T2DM. Aspects analyzed included the presence of mesenchymal stromal cells (MSCs), cellular migration, proliferation, differentiation, apoptosis, and cellular mechanosensitivity. To further verify the computer model findings against in vivo data, an experimental setup was replicated, in which regeneration was compared in healthy and diabetic after a rat femur bone osteotomy stabilized with plate fixation. We found that mechanical alterations had little effect on the reduced bone regeneration in T2DM and that alterations in MSC proliferation, MSC migration, and osteoblast differentiation had the highest effect. In silico predictions of regenerated bone in T2DM matched qualitatively and quantitatively those from ex vivo μCT at 12 weeks post-surgery when reduced cellular activities reported in previous in vitro and in vivo studies were included in the model. The presented findings here could have clinical implications in the treatment of bone fractures in patients with T2DM. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mahdi Jaber
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy AgingTechnische Universität DresdenDresdenGermany
| | - Christine Hofbauer
- Department of Medicine III and Center for Healthy AgingTechnische Universität DresdenDresdenGermany
| | - Georg N Duda
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
- BIH Center for Regenerative TherapiesBIH at Charité ‐ Universitätsmedizin BerlinBerlinGermany
| | - Sara Checa
- Julius Wolff Institute at Berlin Institute of Health, Charité—Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
11
|
Nagrani T, Kumar S, Haq MA, Dhanasekaran S, Gajjar S, Patel C, Sinha S, Haque M. Use of Injectable Platelet-Rich Fibrin Accompanied by Bone Graft in Socket Endurance: A Radiographic and Histological Study. Cureus 2023; 15:e46909. [PMID: 37841989 PMCID: PMC10569439 DOI: 10.7759/cureus.46909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 10/17/2023] Open
Abstract
Background Ridge preservation became a crucial dental health issue and strategy to keep away from ridge defacement after post-tooth loss. The recent scientific evolution of platelet-rich fibrin (PRF) comprises a parenteral formulation of PRF. The combined allograft for socket preservation gives benefits. In this study, bone allografts, demineralized freeze-dried bone allografts (DFDBA) and freeze-dried bone allografts (FDBA) are used in a 30:70 ratio alone or in combination with injectable PRF (I-PRF) for socket preservation. Methods This study is a radiographic and histological examination conducted on 60 participants aged between 19-65 years. Participating patients agreed voluntarily that they would not bear any fixed prosthesis for the next nine months and plan for implanted teeth placement, including multi-rooted mandibular molars denticles. Both groups received atraumatic extraction; then, the socket was preserved with bone allograft alone in the control group and bone allograft mixed with I-PRF, forming sticky bone, in the experimental group. Clinical, radiological, and histological assessments were taken at the inception stage, three months, six months, and nine months. A multivariate regression model and a generalized estimating equation (GEE) model were used to analyse the effects of these changes on outcomes. Results In all the parameters, the test group indicated a good amount of bone growth with increasing intervals of time for bone height radiographically with statistically significant difference present (p<0.05) and histologically after nine months when socket site grafted with bone graft in combination with I-PRF. Conclusion This study's results demonstrated that I-PRF possesses the potential to regenerate and heal in the tooth-extracted socket. This study further recommends the implementation of I-PRF in safeguarding and conserving the raised rim of the tooth. Future research should take place on the osteogenic capability of I-PRF in more comprehensive ridge accession surgical procedures and additional expanding and improving capacities in periodontal reconstruction.
Collapse
Affiliation(s)
- Tanya Nagrani
- Periodontology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Santosh Kumar
- Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Md Ahsanul Haq
- Bio-Statistics, Infectious Diseases Division, icddr, b, Dhaka, BGD
| | | | - Shreya Gajjar
- Periodontology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Chandni Patel
- Periodontology, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Susmita Sinha
- Physiology, Khulna City Medical College and Hospital, Khulna, BGD
| | - Mainul Haque
- Karnavati Scientific Research Center (KSRC), Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
- Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| |
Collapse
|
12
|
Li Y, Meng Y, Wang Y, Wang Y, Wang Z. Application of Mineralized Chitosan Scaffolds in Bone Tissue Engineering. COATINGS 2023; 13:1644. [DOI: 10.3390/coatings13091644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Chitosan (CS) is a natural cationic polysaccharide obtained via the N-deacetylation of chitin. It has various outstanding biological properties such as nontoxicity, biodegradability, biocompatibility, and antimicrobial properties. Minerals can be deposited on the CS template using different methods to construct composites with structures and functions similar to those of natural bone tissue. These ideal scaffolds can produce bone via osteogenesis, osteoinduction, and osteoconduction, with good biocompatibility and mechanical properties, and are thus considered promising novel biomaterials for repairing hard tissue defects. In the last decade, the field of mineralized CS scaffolds has provided novel fundamental knowledge and techniques to better understand the aforementioned fascinating phenomenon. This study mainly focused on the basic structures and properties of mineralized CS scaffolds to understand the current research progress and explore further development. Further, it summarizes the types, preparation methods, components, properties, and applications of mineralized CS scaffolds in bone tissue engineering during the last 5 years. The defects and shortcomings of the scaffolds are discussed, and possible improvement measures are put forward. We aimed to provide complete research progress on mineralized CS scaffolds in bone tissue engineering for researchers and clinicians, and also ideas for the next generation of mineralized CS scaffolds.
Collapse
Affiliation(s)
- Yiyuan Li
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Yufeng Meng
- Division of Nanomaterials & Chemistry Hefei National Laboratory for Physical Sciences at the Microscale Institute of Energy, Hefei Comprehensive National Science Center, CAS Center for Excellence in Nanoscience, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials University of Science and Technology of China, Hefei 230026, China
| | - Yuning Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Yun Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Zuolin Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| |
Collapse
|
13
|
Lin S, Maekawa H, Moeinzadeh S, Lui E, Alizadeh HV, Li J, Kim S, Poland M, Gadomski BC, Easley JT, Young J, Gardner M, Mohler D, Maloney WJ, Yang YP. An osteoinductive and biodegradable intramedullary implant accelerates bone healing and mitigates complications of bone transport in male rats. Nat Commun 2023; 14:4455. [PMID: 37488113 PMCID: PMC10366099 DOI: 10.1038/s41467-023-40149-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Bone transport is a surgery-driven procedure for the treatment of large bone defects. However, challenging complications include prolonged consolidation, docking site nonunion and pin tract infection. Here, we develop an osteoinductive and biodegradable intramedullary implant by a hybrid tissue engineering construct technique to enable sustained delivery of bone morphogenetic protein-2 as an adjunctive therapy. In a male rat bone transport model, the eluting bone morphogenetic protein-2 from the implants accelerates bone formation and remodeling, leading to early bony fusion as shown by imaging, mechanical testing, histological analysis, and microarray assays. Moreover, no pin tract infection but tight osseointegration are observed. In contrast, conventional treatments show higher proportion of docking site nonunion and pin tract infection. The findings of this study demonstrate that the novel intramedullary implant holds great promise for advancing bone transport techniques by promoting bone regeneration and reducing complications in the treatment of bone defects.
Collapse
Affiliation(s)
- Sien Lin
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Hirotsugu Maekawa
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Mechanical Engineering, School of Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Hossein Vahid Alizadeh
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Jiannan Li
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Sungwoo Kim
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Michael Poland
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Benjamin C Gadomski
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jeremiah T Easley
- Preclinical Surgical Research Laboratory, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jeffrey Young
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Michael Gardner
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - David Mohler
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - William J Maloney
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Materials Science and Engineering, School of Engineering, Stanford University, Stanford, CA, 94305, USA.
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Chan ME, Strait L, Ashdown C, Pasumarthy S, Hassan A, Crimarco S, Singh C, Patel VS, Pagnotti G, Khan O, Uzer G, Rubin CT. Low intensity mechanical signals promote proliferation in a cell-specific manner: Tailoring a non-drug strategy to enhance biomanufacturing yields. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547864. [PMID: 37461507 PMCID: PMC10350023 DOI: 10.1101/2023.07.05.547864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Biomanufacturing relies on living cells to produce biotechnology-based therapeutics, tissue engineering constructs, vaccines, and a vast range of agricultural and industrial products. With the escalating demand for these bio-based products, any process that could improve yields and shorten outcome timelines by accelerating cell proliferation would have a significant impact across the discipline. While these goals are primarily achieved using biological or chemical strategies, harnessing cell mechanosensitivity represents a promising - albeit less studied - physical pathway to promote bioprocessing endpoints, yet identifying which mechanical parameters influence cell activities has remained elusive. We tested the hypothesis that mechanical signals, delivered non-invasively using low-intensity vibration (LIV; <1g, 10-500Hz), will enhance cell expansion, and determined that any unique signal configuration was not equally influential across a range of cell types. Varying frequency, intensity, duration, refractory period, and daily doses of LIV increased proliferation in CHO-adherent cells (+79% in 96h) using a particular set of LIV parameters (0.2g, 500Hz, 3x30 min/d, 2h refractory period), yet this same mechanical input suppressed proliferation in CHO-suspension cells (-13%). Exposing these same CHO-suspension cells to distinct LIV parameters (30Hz, 0.7g, 2x60 min/d, 2h refractory period) increased proliferation by 210%. Particle image velocimetry combined with finite element modeling showed high transmissibility of these signals across fluids (>90%), and LIV effectively scaled up to T75 flasks. Ultimately, when LIV is tailored to the target cell population, its highly efficient transmission across media represents a means to non-invasively augment biomanufacturing endpoints for both adherent and suspended cells, and holds immediate applications, ranging from small-scale, patient-specific personalized medicine to large-scale commercial bio-centric production challenges.
Collapse
|
15
|
Patel K, Mangu SR, Sukhdeo SV, Sharan K. Sesamol improves bone mass in ovary intact growing and adult rats but accelerates bone deterioration in the ovariectomized rats. J Nutr Biochem 2023:109384. [PMID: 37209954 DOI: 10.1016/j.jnutbio.2023.109384] [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: 12/16/2022] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Sesamol, an active component in sesame seeds, is known for its health benefits. However, its effect on bone metabolism remains unexplored. The present study aims to investigate the effect of sesamol on growing, adult and osteoporotic skeleton and its mechanism of action. Sesamol at various doses were administered orally to growing, ovariectomized, and ovary-intact rats. Alterations in bone parameters were examined using micro-CT and histological studies. Western blot and mRNA expression from long bones were performed. We further evaluated the effect of sesamol on osteoblast and osteoclast function and its mode of action in the cell culture system. These data showed that sesamol was able to promote peak bone mass in growing rats. However, sesamol had the opposite effect in ovariectomized rats, evident from gross deterioration of trabecular and cortical microarchitecture. Concurrently, it improved the bone mass in adult rats. In vitro results revealed that sesamol enhances the bone formation by stimulating osteoblast differentiation through MAPK, AKT, and BMP-2 signaling. In contrast, it enhances osteoclast differentiation and expression of osteoclast-specific genes in osteoclast differentiation medium. Interestingly, in presence of estrogen, the effect reversed and sesamol decreased osteoclast differentiation, in vitro. Sesamol improves bone microarchitecture in growing and ovary-intact rats, whereas it enhances the bone deterioration in ovariectomized rats. While sesamol promotes bone formation, its opposing effect on the skeleton can be attributed to its dual effect on osteoclastogenesis in presence and absence of estrogen. These findings in the preclinical context suggests a special attention towards the detrimental effect of sesamol in postmenopausal women.
Collapse
Affiliation(s)
- Kalpana Patel
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Svvs Ravi Mangu
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shinde Vijay Sukhdeo
- Department of Meat and Marine Sciences, CSIR- Central Food Technological Research Institute, Mysuru, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
16
|
Wang YW, Lin WY, Wu FJ, Luo CW. Unveiling the transcriptomic landscape and the potential antagonist feedback mechanisms of TGF-β superfamily signaling module in bone and osteoporosis. Cell Commun Signal 2022; 20:190. [PMID: 36443839 PMCID: PMC9703672 DOI: 10.1186/s12964-022-01002-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/22/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND TGF-β superfamily signaling is indispensable for bone homeostasis. However, the global expression profiles of all the genes that make up this signaling module in bone and bone-related diseases have not yet been well characterized. METHODS Transcriptomic datasets from human bone marrows, bone marrow-derived mesenchymal stem cells (MSCs) and MSCs of primary osteoporotic patients were used for expression profile analyses. Protein treatments, gene quantification, reporter assay and signaling dissection in MSC lines were used to clarify the interactive regulations and feedback mechanisms between TGF-β superfamily ligands and antagonists. Ingenuity Pathway Analysis was used for network construction. RESULTS We identified TGFB1 in the ligand group that carries out SMAD2/3 signaling and BMP8A, BMP8B and BMP2 in the ligand group that conducts SMAD1/5/8 signaling have relatively high expression levels in normal bone marrows and MSCs. Among 16 antagonist genes, the dominantly expressed TGF-β superfamily ligands induced only NOG, GREM1 and GREM2 via different SMAD pathways in MSCs. These induced antagonist proteins further showed distinct antagonisms to the treated ligands and thus would make up complicated negative feedback networks in bone. We further identified TGF-β superfamily signaling is enriched in MSCs of primary osteoporosis. Enhanced expression of the genes mediating TGF-β-mediated SMAD3 signaling and the genes encoding TGF-β superfamily antagonists served as significant features to osteoporosis. CONCLUSION Our data for the first time unveiled the transcription landscape of all the genes that make up TGF-β superfamily signaling module in bone. The feedback mechanisms and regulatory network prediction of antagonists provided novel hints to treat osteoporosis. Video Abstract.
Collapse
Affiliation(s)
- Ying-Wen Wang
- grid.260539.b0000 0001 2059 7017Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112 Taiwan
| | - Wen-Yu Lin
- grid.260539.b0000 0001 2059 7017Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112 Taiwan
| | - Fang-Ju Wu
- grid.260539.b0000 0001 2059 7017Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112 Taiwan
| | - Ching-Wei Luo
- grid.260539.b0000 0001 2059 7017Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112 Taiwan
| |
Collapse
|
17
|
Park KR, Park JI, Lee S, Yoo K, Kweon GR, Kwon IK, Yun HM, Hong JT. Chi3L1 is a therapeutic target in bone metabolism and a potential clinical marker in patients with osteoporosis. Pharmacol Res 2022; 184:106423. [PMID: 36064078 DOI: 10.1016/j.phrs.2022.106423] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 01/03/2023]
Abstract
BMP2 is clinically used as an ectopic bone inducer and plays a significant role in bone development, formation, and diseases. Chitinase 3-like 1 protein (Chi3L1) is found in the skeletal system. However, Chi3L1-mediated bone metabolism and aging-related bone erosion via BMP2 signaling have not yet been demonstrated. Herein, Chi3L1 increased BMP2-induced osteoblast differentiation in mesenchymal precursor cells and human primary osteoblasts. Chi3L1KO(-/-) showed abnormal bone development, and primary osteoblasts isolated from Chi3L1KO(-/-) exhibited impaired osteoblast differentiation and maturation. Chi3L1 also potentiated BMP2 signaling and RUNX2 expression in primary osteoblasts. Chi3L1 interacted with BMPRIa, which increased the surface expression of BMPRIa and promoted BMP2 signaling to induce osteoblast differentiation. Chi3L1KO(-/-) mice showed bone formation reduced with a decrease in RUNX2 expression in calvarial defects. Chi3L1KO(-/-) mice exhibited aging-related osteoporotic bone loss with decreases in the levels of RUNX2 and OPG, while serum PYD level and osteoclast number increased. Chi3L1 increased OPG via non-canonical BMP2 signaling in osteoblasts, which suppressed osteoclastogenesis in BMMs. Furthermore, ROC analysis showed that serum Chi3L1 level clinically decreased in osteoporosis patients. Our findings demonstrate that Chi3L1 promotes bone formation, suppresses osteoclastogenesis, and prevents aging-related osteoporosis.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea.
| | - Jae-Il Park
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61751, Republic of Korea.
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea.
| | - Kyeongwon Yoo
- KRIBB/Bio-venture Center, Daejeon 34141, Republic of Korea.
| | - Gi-Ryang Kweon
- Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Chungnam 34134, Republic of Korea.
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02453, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
18
|
Howard MT, Wang S, Berger AG, Martin JR, Jalili-Firoozinezhad S, Padera RF, Hammond PT. Sustained release of BMP-2 using self-assembled layer-by-layer film-coated implants enhances bone regeneration over burst release. Biomaterials 2022; 288:121721. [PMID: 35981926 PMCID: PMC10396073 DOI: 10.1016/j.biomaterials.2022.121721] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
Abstract
Current clinical products delivering the osteogenic growth factor bone morphogenetic protein 2 (BMP-2) for bone regeneration have been plagued by safety concerns due to a high incidence of off-target effects resulting from bolus release and supraphysiological doses. Layer-by-layer (LbL) film deposition offers the opportunity to coat bone defect-relevant substrates with thin films containing proteins and other therapeutics; however, control of release kinetics is often hampered by interlayer diffusion of drugs throughout the film during assembly, which causes burst drug release. In this work, we present the design of different laponite clay diffusional barrier layer architectures in self-assembled LbL films to modulate the release kinetics of BMP-2 from the surface of a biodegradable implant. Release kinetics were tuned by incorporating laponite in different film arrangements and with varying deposition techniques to achieve release of BMP-2 over 2 days, 4 days, 14 days, and 30 days. Delivery of a low dose (0.5 μg) of BMP-2 over 2 days and 30 days using these LbL film architectures was then compared in an in vivo rat critical size calvarial defect model to determine the effect of BMP-2 release kinetics on bone regeneration. After 6 weeks, sustained release of BMP-2 over 30 days induced 3.7 times higher bone volume and 7.4 times higher bone mineral density as compared with 2-day release of BMP-2, which did not induce more bone growth than the uncoated scaffold control. These findings represent a crucial step in the understanding of how BMP-2 release kinetics influence treatment efficacy and underscore the necessity to optimize protein delivery methods in clinical formulations for bone regeneration. This work could be applied to the delivery of other therapeutic proteins for which careful tuning of the release rate is a key optimization parameter.
Collapse
Affiliation(s)
- MayLin T Howard
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sheryl Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Adam G Berger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - John R Martin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Robert F Padera
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, United States.
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| |
Collapse
|
19
|
Babaei M, Nasernejad B, Sharifikolouei E, Shokrgozar MA, Bonakdar S. Bioactivation of 3D Cell-Imprinted Polydimethylsiloxane Surfaces by Bone Protein Nanocoating for Bone Tissue Engineering. ACS OMEGA 2022; 7:26353-26367. [PMID: 35936447 PMCID: PMC9352215 DOI: 10.1021/acsomega.2c02206] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/04/2022] [Indexed: 06/03/2023]
Abstract
Physical and chemical parameters that mimic the physiological niche of the human body have an influence on stem cell fate by creating directional signals to cells. Micro/nano cell-patterned polydimethylsiloxane (PDMS) substrates, due to their ability to mimic the physiological niche, have been widely used in surface modification. Integration of other factors such as the biochemical coating on the surface can achieve more similar microenvironmental conditions and promote stem cell differentiation to the target cell line. Herein, we investigated the effect of physical topography, chemical functionalization by acid bone lysate (ABL) nanocoating, and the combined functionalization of the bone proteins' nanocoated surface and the topographically modified surface. We prepared four distinguishing surfaces: plain PDMS, physically modified PDMS by 3D cell topography patterning, chemically modified PDMS with bone protein nanocoating, and chemically modified nano 3D cell-imprinted PDMS by bone proteins (ABL). Characterization of extracted ABL was carried out by Bradford staining and sodium dodecyl sulfate polyacrylamide gel electrophoresis analysis, followed by the MTT assay for evaluation of cell viability on ABL-coated PDMS. Moreover, field emission scanning electron microscopy and profilometry were used for the determination of optimal coating thickness, and the appropriate coating concentration was identified and used in the study. The binding and retention of ABL to PDMS were confirmed by Fourier transform infrared spectroscopy and bicinchoninic acid assay. Sessile drop static water contact angle measurements on substrates showed that the combined chemical functionalization and nano 3D cell-imprinting on the PDMS surface improved surface wettability by 66% compared to plain PDMS. The results of ALP measurement, alizarin red S staining, immunofluorescence staining, and real-time PCR showed that the nano 3D cell-imprinted PDMS surface functionalized by extracted bone proteins, ABL, is able to guide the fate of adipose derived stem cellss toward osteogenic differentiation. Eventually, chemical modification of the cell-imprinted PDMS substrate by bone protein extraction not only improved the cell adhesion and proliferation but also contributed to the topographical effect itself and caused a significant synergistic influence on the process of osteogenic differentiation.
Collapse
Affiliation(s)
- Mahrokh Babaei
- Department
of Chemical Engineering, Amirkabir University
of Technology (Tehran Polytechnic), Tehran 15875-4413, Iran
| | - Bahram Nasernejad
- Department
of Chemical Engineering, Amirkabir University
of Technology (Tehran Polytechnic), Tehran 15875-4413, Iran
| | - Elham Sharifikolouei
- Department
of Applied Science and Technology, Politecnico
di Torino, Turin 10129, Italy
| | | | - Shahin Bonakdar
- National
Cell Bank, Pasteur Institute of Iran, Tehran 13169-43551, Iran
| |
Collapse
|
20
|
Walther AR, Ditzel N, Kassem M, Andersen MØ, Hedegaard MAB. In vivo non-invasive monitoring of tissue development in 3D printed subcutaneous bone scaffolds using fibre-optic Raman spectroscopy. BIOMATERIALS AND BIOSYSTEMS 2022; 7:100059. [PMID: 36824488 PMCID: PMC9934492 DOI: 10.1016/j.bbiosy.2022.100059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022] Open
Abstract
The development of novel biomaterials for regenerative therapy relies on the ability to assess tissue development, quality, and similarity with native tissue types in in vivo experiments. Non-invasive imaging modalities such as X-ray computed tomography offer high spatial resolution but limited biochemical information while histology and biochemical assays are destructive. Raman spectroscopy is a non-invasive, label-free and non-destructive technique widely applied for biochemical characterization. Here we demonstrate the use of fibre-optic Raman spectroscopy for in vivo quantitative monitoring of tissue development in subcutaneous calcium phosphate scaffolds in mice over 16 weeks. Raman spectroscopy was able to quantify the time dependency of different tissue components related to the presence, absence, and quantity of mesenchymal stem cells. Scaffolds seeded with stem cells produced 3-5 times higher amount of collagen-rich extracellular matrix after 16 weeks implantation compared to scaffolds without. These however, showed a 2.5 times higher amount of lipid-rich tissue compared to implants with stem cells. Ex vivo micro-computed tomography and histology showed stem cell mediated collagen and bone development. Histological measures of collagen correlated well with Raman derived quantifications (correlation coefficient in vivo 0.74, ex vivo 0.93). In the absence of stem cells, the scaffolds were largely occupied by adipocytes. The technique developed here could potentially be adapted for a range of small animal experiments for assessing tissue engineering strategies at the biochemical level.
Collapse
Affiliation(s)
- Anders Runge Walther
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Nicholas Ditzel
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Moustapha Kassem
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Morten Østergaard Andersen
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Martin Aage Barsøe Hedegaard
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
21
|
Long-term posterolateral spinal fusion in rabbits induced by rhBMP6 applied in autologous blood coagulum with synthetic ceramics. Sci Rep 2022; 12:11649. [PMID: 35803983 PMCID: PMC9270325 DOI: 10.1038/s41598-022-14931-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Autologous bone graft substitute (ABGS) containing rhBMP6 in autologous blood coagulum (Osteogrow) is a novel therapeutic solution for bone regeneration. This study is aimed to investigate the long-term outcome of ABGS with synthetic ceramics (Osteogrow-C) in rabbit posterolateral spinal fusion (PLF) model. Osteogrow-C implants were implanted bilaterally between rabbit lumbar transverse processes. We compared the outcome following implantation of ABGS with ceramic particles of different chemical composition (TCP and biphasic ceramics containing both TCP and HA) and size (500–1700 µm and 74–420 µm). Outcome was analyzed after 14 and 27 weeks by microCT, histology, and biomechanical analyses. Successful bilateral spinal fusion was observed in all animals at the end of observation period. Chemical composition of ceramic particles has impact on the PLF outcome via resorption of TCP ceramics, while ceramics containing HA were only partially resorbed. Moreover, persistence of ceramic particles subsequently resulted with an increased bone volume in implants with small particles containing high proportion of HA. ABGS (rhBMP6/ABC) with various synthetic ceramic particles promoted spinal fusion in rabbits. This is the first presentation of BMP-mediated ectopic bone formation in rabbit PLF model with radiological, histological, and biomechanical features over a time course of up to 27 weeks.
Collapse
|
22
|
Khan MM, Serajuddin M, Malik MZ. Identification of microRNA and gene interactions through bioinformatic integrative analysis for revealing candidate signatures in prostate cancer. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
23
|
Szilágyi SS, Amsalem-Zafran AR, Shapira KE, Ehrlich M, Henis YI. Competition between type I activin and BMP receptors for binding to ACVR2A regulates signaling to distinct Smad pathways. BMC Biol 2022; 20:50. [PMID: 35177083 PMCID: PMC8855587 DOI: 10.1186/s12915-022-01252-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 02/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Activins and bone morphogenetic proteins (BMPs) play critical, sometimes opposing roles, in multiple physiological and pathological processes and diseases. They signal to distinct Smad branches; activins signal mainly to Smad2/3, while BMPs activate mainly Smad1/5/8. This gives rise to the possibility that competition between the different type I receptors through which activin and BMP signal for common type II receptors can provide a mechanism for fine-tuning the cellular response to activin/BMP stimuli. Among the transforming growth factor-β superfamily type II receptors, ACVR2A/B are highly promiscuous, due to their ability to interact with different type I receptors (e.g., ALK4 vs. ALK2/3/6) and with their respective ligands [activin A (ActA) vs. BMP9/2]. However, studies on complex formation between these full-length receptors situated at the plasma membrane, and especially on the potential competition between the different activin and BMP type I receptors for a common activin type II receptor, were lacking. Results We employed a combination of IgG-mediated patching-immobilization of several type I receptors in the absence or presence of ligands with fluorescence recovery after photobleaching (FRAP) measurements on the lateral diffusion of an activin type II receptor, ACVR2A, to demonstrate the principle of competition between type I receptors for ACVR2. Our results show that ACVR2A can form stable heteromeric complexes with ALK4 (an activin type I receptor), as well as with several BMP type I receptors (ALK2/3/6). Of note, ALK4 and the BMP type I receptors competed for binding ACVR2A. To assess the implications of this competition for signaling output, we first validated that in our cell model system (U2OS cells), ACVR2/ALK4 transduce ActA signaling to Smad2/3, while BMP9 signaling to Smad1/5/8 employ ACVR2/ALK2 or ACVR2/ALK3. By combining ligand stimulation with overexpression of a competing type I receptor, we showed that differential complex formation of distinct type I receptors with a common type II receptor balances the signaling to the two Smad branches. Conclusions Different type I receptors that signal to distinct Smad pathways (Smad2/3 vs. Smad1/5/8) compete for binding to common activin type II receptors. This provides a novel mechanism to balance signaling between Smad2/3 and Smad1/5/8. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01252-z.
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Ayelet R Amsalem-Zafran
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Keren E Shapira
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
24
|
Pinter ZW, Elder BD, Kaye ID, Kepler CK, Wagner S, Freedman BA, Sebastian AS. A Review of Commercially Available Cellular-based Allografts. Clin Spine Surg 2022; 35:E77-E86. [PMID: 34654775 DOI: 10.1097/bsd.0000000000001262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/15/2021] [Indexed: 11/27/2022]
Abstract
STUDY DESIGN This was a narrative review. OBJECTIVE This review discusses our current knowledge regarding cellular-based allografts while highlighting the key gaps in the literature that must be addressed before their widespread adoption. SUMMARY OF BACKGROUND DATA Iliac crest bone graft is the gold-standard bone graft material but is associated with donor site morbidity. Commonly utilized bone graft extenders such as demineralized bone matrix and bone morphogenetic protein have conflicting data supporting their efficacy and lack the osteogenic potential of new cellular-based allograft options. METHODS An extensive literature review was performed. The literature was then summarized in accordance with the authors' clinical experience. RESULTS There is not widespread evidence thus far that the addition of the osteogenic cellular component to allograft enhances spinal fusion, as a recent study by Bhamb and colleagues demonstrated superior bone formation during spine fusion in an aythmic rat model when demineralized bone matrix was used in comparison to Osteocel Plus. Furthermore, the postimplantation cellular viability and osteogenic and osteoinductive capacity of cellular-based allografts need to be definitively established, especially given that a recent study by Lina and colleagues demonstrated a paucity of bone marrow cell survival in an immunocompetent mouse posterolateral spinal fusion model. CONCLUSIONS This data indicates that the substantially increased cost of these cellular allografts may not be justified. LEVEL OF EVIDENCE Level V.
Collapse
Affiliation(s)
| | | | - I David Kaye
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA
| | | | - Scott Wagner
- Walter Reed National Military Medical Center, Bethesda, MD
| | | | | |
Collapse
|
25
|
Zhang X, Li J, Chen J, Peng Z, Chen J, Liu X, Wu F, Zhang P, Chen GGQ. Enhanced Bone Regeneration via PHA Scaffolds Coated with Polydopamine-Captured BMP2. J Mater Chem B 2022; 10:6214-6227. [DOI: 10.1039/d2tb01122k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The hierarchical three-dimensional (3D)-printing scaffolds based on microbial polyester poly(3-hydrxoybutyrate-co-4-hydroxybutyrate) (P34HB) were designed and used for bone tissue engineering via surface functionalization on the 3D-printed (P34HB) scaffolds using polydopamine (PDA)-mediated...
Collapse
|
26
|
Osteogenic Competence and Potency of the Bone Induction Principle: Inductive Substrates That Initiate “Bone: Formation by Autoinduction”. J Craniofac Surg 2021; 33:971-984. [DOI: 10.1097/scs.0000000000008299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
27
|
Zhang X, Fan J, Chen C, Aghaloo T, Lee M. Co-delivery of simvastatin and demineralized bone matrix hierarchically from nanosheet-based supramolecular hydrogels for osteogenesis. J Mater Chem B 2021; 9:7741-7750. [PMID: 34586142 DOI: 10.1039/d1tb01256h] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Supramolecular hydrogels are widely used as 3D scaffolds and delivery platforms in tissue engineering applications. However, hydrophobic therapeutic agents exhibit weak compatibility in hydrogel scaffolds along with aggregation and precipitation. Herein, simvastatin drugs used as BMP-2 stimulators are encapsulated into the layer space of LAPONITE® via electrostatic interactions and ion exchange efficiently, and supramolecular hydrogels could be fabricated with a self-healing, injectable and sustained drug release nature. Hydrogels encapsulated with 10 μg mL-1 simvastatin drug show good osteogenic differentiation in vitro. Moreover, the loading of demineralized bone matrix particles could enhance the capacity for osteogenesis via improving the expression of BMP-2 synergistically. The integrated hydrogels could be implanted into cranial defect sites for bone regeneration in vivo. This work provides the first demonstration of molecular and supramolecular engineering of hydrogels to load osteoinductive agents hierarchically for bone regeneration, contributing to the development of a brand-new strategy for dealing with compatibility between scaffolds and osteogenic agents.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pharmacy, Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi 030001, P. R. China. .,Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA.
| | - Jiabing Fan
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA.
| | - Chen Chen
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA.
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA. .,Department of Bioengineering, University of California at Los Angeles, 420 Westwood Plaza, Los Angeles, California 90095, USA
| |
Collapse
|
28
|
Chondrogenic Potential of Human Dental Pulp Stem Cells Cultured as Microtissues. Stem Cells Int 2021; 2021:7843798. [PMID: 34539791 PMCID: PMC8443354 DOI: 10.1155/2021/7843798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/22/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
Several tissue engineering stem cell-based procedures improve hyaline cartilage repair. In this work, the chondrogenic potential of dental pulp stem cell (DPSC) organoids or microtissues was studied. After several weeks of culture in proliferation or chondrogenic differentiation media, synthesis of aggrecan and type II and I collagen was immunodetected, and SOX9, ACAN, COL2A1, and COL1A1 gene expression was analysed by real-time RT-PCR. Whereas microtissues cultured in proliferation medium showed the synthesis of aggrecan and type II and I collagen at the 6th week of culture, samples cultured in chondrogenic differentiation medium showed an earlier and important increase in the synthesis of these macromolecules after 4 weeks. Gene expression analysis showed a significant increase of COL2A1 after 3 days of culture in chondrogenic differentiation medium, while COL1A1 was highly expressed after 14 days. Cell-cell proximity promotes the chondrogenic differentiation of DPSCs and important synthesis of hyaline chondral macromolecules.
Collapse
|
29
|
Divya D, Bhattacharya TK. Bone morphogenetic proteins (BMPs) and their role in poultry. WORLD POULTRY SCI J 2021. [DOI: 10.1080/00439339.2021.1959274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- D. Divya
- Molecular Genetics and Breeding Division, ICAR-Directorate of Poultry Research, Hyderabad, India
| | - T. K. Bhattacharya
- Molecular Genetics and Breeding Division, ICAR-Directorate of Poultry Research, Hyderabad, India
| |
Collapse
|
30
|
Tavafoghi M, Darabi MA, Mahmoodi M, Tutar R, Xu C, Mirjafari A, Billi F, Swieszkowski W, Nasrollahi F, Ahadian S, Hosseini V, Khademhosseini A, Ashammakhi N. Multimaterial bioprinting and combination of processing techniques towards the fabrication of biomimetic tissues and organs. Biofabrication 2021; 13. [PMID: 34130266 DOI: 10.1088/1758-5090/ac0b9a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022]
Abstract
Tissue reconstruction requires the utilization of multiple biomaterials and cell types to replicate the delicate and complex structure of native tissues. Various three-dimensional (3D) bioprinting techniques have been developed to fabricate customized tissue structures; however, there are still significant challenges, such as vascularization, mechanical stability of printed constructs, and fabrication of gradient structures to be addressed for the creation of biomimetic and complex tissue constructs. One approach to address these challenges is to develop multimaterial 3D bioprinting techniques that can integrate various types of biomaterials and bioprinting capabilities towards the fabrication of more complex structures. Notable examples include multi-nozzle, coaxial, and microfluidics-assisted multimaterial 3D bioprinting techniques. More advanced multimaterial 3D printing techniques are emerging, and new areas in this niche technology are rapidly evolving. In this review, we briefly introduce the basics of individual 3D bioprinting techniques and then discuss the multimaterial 3D printing techniques that can be developed based on combination of these techniques for the engineering of complex and biomimetic tissue constructs. We also discuss the perspectives and future directions to develop state-of-the-art multimaterial 3D bioprinting techniques for engineering tissues and organs.
Collapse
Affiliation(s)
- Maryam Tavafoghi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Mahboobeh Mahmoodi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Rumeysa Tutar
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa Avcılar, Istanbul 34320, Turkey
| | - Chun Xu
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,School of Dentistry, The University of Queensland, Brisbane, Australia
| | - Arshia Mirjafari
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Fabrizio Billi
- UCLA/OIC Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States of America
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Fatemeh Nasrollahi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Vahid Hosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America.,Department of Chemical Engineering, University of California, Los Angeles, CA, United States of America
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, College of Engineering, Michigan State University, MI, United States of America
| |
Collapse
|
31
|
A Cyclic BMP-2 Peptide Upregulates BMP-2 Protein-Induced Cell Signaling in Myogenic Cells. Polymers (Basel) 2021; 13:polym13152549. [PMID: 34372154 PMCID: PMC8347162 DOI: 10.3390/polym13152549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022] Open
Abstract
In the current study, we designed four cyclic peptide analogues by incorporating two cysteine residues in a BMP-2 linear knuckle epitope in such a way that the active region of the peptide could be either inside or outside the cyclic ring. Bone morphogenetic protein receptor BMPRII was immobilized on the chip surface, and the interaction of the linear and cyclic peptide analogues was studied using surface plasmon resonance (SPR). From the affinity data, the peptides with an active region inside the cyclic ring had a higher binding affinity in comparison to the other peptides. To confirm that our affinity data are in line in vitro, we studied the expression levels of RUNX2 (runt-related transcription factor) and conducted an osteogenic marker alkaline phosphatase (ALP) assay and staining. Based on the affinity data and the in vitro experiments, peptide P-05 could be a suitable candidate for osteogenesis, with higher binding affinity and increased RUNX2 and ALP expression in comparison to the linear peptides.
Collapse
|
32
|
Borgiani E, Duda GN, Willie BM, Checa S. Bone morphogenetic protein 2-induced cellular chemotaxis drives tissue patterning during critical-sized bone defect healing: an in silico study. Biomech Model Mechanobiol 2021; 20:1627-1644. [PMID: 34047890 PMCID: PMC8298257 DOI: 10.1007/s10237-021-01466-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022]
Abstract
Critical-sized bone defects are critical healing conditions that, if left untreated, often lead to non-unions. To reduce the risk, critical-sized bone defects are often treated with recombinant human BMP-2. Although enhanced bone tissue formation is observed when BMP-2 is administered locally to the defect, spatial and temporal distribution of callus tissue often differs from that found during regular bone healing or in defects treated differently. How this altered tissue patterning due to BMP-2 treatment is linked to mechano-biological principles at the cellular scale remains largely unknown. In this study, the mechano-biological regulation of BMP-2-treated critical-sized bone defect healing was investigated using a multiphysics multiscale in silico approach. Finite element and agent-based modeling techniques were combined to simulate healing within a critical-sized bone defect (5 mm) in a rat femur. Computer model predictions were compared to in vivo microCT data outcome of bone tissue patterning at 2, 4, and 6 weeks postoperation. In vivo, BMP-2 treatment led to complete healing through periosteal bone bridging already after 2 weeks postoperation. Computer model simulations showed that the BMP-2 specific tissue patterning can be explained by the migration of mesenchymal stromal cells to regions with a specific concentration of BMP-2 (chemotaxis). This study shows how computational modeling can help us to further understand the mechanisms behind treatment effects on compromised healing conditions as well as to optimize future treatment strategies.
Collapse
Affiliation(s)
- Edoardo Borgiani
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bettina M Willie
- Research Centre, Department of Pediatric Surgery, Shriners Hospital for Children-Canada, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Sara Checa
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
33
|
Shirazi S, Huang CC, Kang M, Lu Y, Ravindran S, Cooper LF. The importance of cellular and exosomal miRNAs in mesenchymal stem cell osteoblastic differentiation. Sci Rep 2021; 11:5953. [PMID: 33723364 PMCID: PMC7960990 DOI: 10.1038/s41598-021-85306-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The differentiation of osteoblasts is under complex regulation that includes autocrine and paracrine signaling from MSCs. Exosomes are important components of the MSC secretome and their cargo contains numerous miRNAs. In this study, the importance of MSC miRNAs in modulation of osteoblastic differentiation was examined by global reduction of miRNA biosynthesis in Dicer knock down hMSCs. We additionally impaired hMSC responses to miRNAs by knockdown of Argonaute 2 expression. Knockdown of Dicer and Argonaute 2 both reduced osteoblastic differentiation of hMSCs. This was observed at the levels of hMSC culture mineralization and osteoblastic gene expression. The treatment of Dicer deficient hMSCs with wild type hMSC exosomes effectively recovered the impaired osteoblastic differentiation. Dicer knockdown reduced the quantity and diversity of miRNAs present in hMSC exosomes. miRSeq data and KEGG analysis implicated the miRNA-dependent effects on multiple osteoinductive pathways in Dicer deficient cells, including the Hippo signaling and TGF-beta signaling pathways. Treatment of hMSCs with mimics of miRNAs significantly downregulated in Dicer knockdown cells recovered functions of exosome-mediated signaling in hMSCs. These results indicate that hMSC exosomes exert miRNA-dependent control that contributes to osteoblastic differentiation.
Collapse
Affiliation(s)
- Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Chun-Chieh Huang
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Miya Kang
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Yu Lu
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA.
| | - Lyndon F Cooper
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Room 561C, Chicago, IL, 60612, USA.
| |
Collapse
|
34
|
Amarasekara DS, Kim S, Rho J. Regulation of Osteoblast Differentiation by Cytokine Networks. Int J Mol Sci 2021; 22:ijms22062851. [PMID: 33799644 PMCID: PMC7998677 DOI: 10.3390/ijms22062851] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoblasts, which are bone-forming cells, play pivotal roles in bone modeling and remodeling. Osteoblast differentiation, also known as osteoblastogenesis, is orchestrated by transcription factors, such as runt-related transcription factor 1/2, osterix, activating transcription factor 4, special AT-rich sequence-binding protein 2 and activator protein-1. Osteoblastogenesis is regulated by a network of cytokines under physiological and pathophysiological conditions. Osteoblastogenic cytokines, such as interleukin-10 (IL-10), IL-11, IL-18, interferon-γ (IFN-γ), cardiotrophin-1 and oncostatin M, promote osteoblastogenesis, whereas anti-osteoblastogenic cytokines, such as tumor necrosis factor-α (TNF-α), TNF-β, IL-1α, IL-4, IL-7, IL-12, IL-13, IL-23, IFN-α, IFN-β, leukemia inhibitory factor, cardiotrophin-like cytokine, and ciliary neurotrophic factor, downregulate osteoblastogenesis. Although there are gaps in the body of knowledge regarding the interplay of cytokine networks in osteoblastogenesis, cytokines appear to be potential therapeutic targets in bone-related diseases. Thus, in this study, we review and discuss our osteoblast, osteoblast differentiation, osteoblastogenesis, cytokines, signaling pathway of cytokine networks in osteoblastogenesis.
Collapse
Affiliation(s)
- Dulshara Sachini Amarasekara
- Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo 00300, Sri Lanka;
| | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea;
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea;
- Correspondence: ; Tel.: +82-42-821-6420; Fax: +82-42-822-7367
| |
Collapse
|
35
|
Pauk M, Kufner V, Rumenovic V, Dumic-Cule I, Farkas V, Milosevic M, Bordukalo-Niksic T, Vukicevic S. Iron overload in aging Bmp6‑/‑ mice induces exocrine pancreatic injury and fibrosis due to acinar cell loss. Int J Mol Med 2021; 47:60. [PMID: 33649802 PMCID: PMC7910010 DOI: 10.3892/ijmm.2021.4893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 01/19/2021] [Indexed: 11/27/2022] Open
Abstract
The relationship between hemochromatosis and diabetes has been well established, as excessive iron deposition has been reported to result in impaired function of the endocrine and exocrine pancreas. Therefore, the objective of the present study was to analyze the effects of iron accumulation on the pancreata and glucose homeostasis in a bone morphogenetic protein 6-knockout (Bmp6−/−) mouse model of hemochromatosis. The sera and pancreatic tissues of wild-type (WT) and Bmp6−/− mice (age, 3 and 10 months) were subjected to biochemical and histological analyses. In addition, 18F-fluorodeoxyglucose biodistribution was evaluated in the liver, muscle, heart, kidney and adipose tissue of both animal groups. The results demonstrated that 3-month-old Bmp6−/− mice exhibited iron accumulation preferentially in the exocrine pancreas, with no signs of pancreatic injury or fibrosis. No changes were observed in the glucose metabolism, as pancreatic islet diameter, insulin and glucagon secretion, blood glucose levels and glucose uptake in the liver, muscle and adipose tissue remained comparable with those in the WT mice. Aging Bmp6−/− mice presented with progressive iron deposits in the exocrine pancreas, leading to pancreatic degeneration and injury that was characterized by acinar atrophy, fibrosis and the infiltration of inflammatory cells. However, the aging mice exhibited unaltered blood glucose levels and islet structure, normal insulin secretion and moderately increased α-cell mass compared with those in the age-matched WT mice. Additionally, iron overload and pancreatic damage were not observed in the aging WT mice. These results supported a pathogenic role of iron overload in aging Bmp6−/− mice leading to iron-induced exocrine pancreatic deficiency, whereas the endocrine pancreas retained normal function.
Collapse
Affiliation(s)
- Martina Pauk
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Vera Kufner
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Viktorija Rumenovic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Ivo Dumic-Cule
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Vladimir Farkas
- Molecular Biology Department, Rudjer Boskovic Institute, HR‑10000 Zagreb, Croatia
| | - Milan Milosevic
- Andrija Stampar School of Public Health, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Tatjana Bordukalo-Niksic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, HR‑10000 Zagreb, Croatia
| |
Collapse
|
36
|
Surface modification of a three-dimensional polycaprolactone scaffold by polydopamine, biomineralization, and BMP-2 immobilization for potential bone tissue applications. Colloids Surf B Biointerfaces 2021; 199:111528. [PMID: 33385823 DOI: 10.1016/j.colsurfb.2020.111528] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/23/2022]
Abstract
Three-dimensional (3D) bioprinting is a free-form fabrication technique enabling fine feature control for tissue engineering applications. Especially, 3D scaffolds capable of supporting cell attachment, proliferation, and osteogenic differentiation are a prerequisite for bone tissue regeneration. Herein, we elaborated this approach to produce a 3D polycaprolactone (PCL) scaffold with long-term osteogenic activity. Specifically, we coated polydopamine (PDA) on 3D PCL scaffolds, subsequently deposited hydroxyapatite (HA) nanoparticles via biomimetic mineralization, and finally immobilized bone morphogenetic protein-2 (BMP-2). Material properties were characterized and compared with various 3D scaffolds, including PCL, PDA-coated PCL (PCL/PDA), and PDA-coated and HA-deposited PCL (PCL/PDA/HA). In vitro cell culture studies with osteoblasts revealed that the PCL/PDA/HA scaffolds immobilized with BMP-2 showed long-term retention of BMP-2 (for up to 21 days) and significantly increased osteoblast proliferation and osteogenic differentiation, as evidenced by metabolic activity, alkaline phosphatase activity, and calcium deposition. We believe that this multifunctional osteogenic 3D scaffold will be useful for bone tissue engineering applications.
Collapse
|
37
|
Sampath TK, Vukicevic S. Biology of bone morphogenetic protein in bone repair and regeneration: A role for autologous blood coagulum as carrier. Bone 2020; 141:115602. [PMID: 32841742 DOI: 10.1016/j.bone.2020.115602] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022]
Abstract
BMPs were purified from demineralized bone matrix based on their ability to induce new bone in vivo and they represent a large member of the TGF-β superfamily of proteins. BMPs serve as morphogenic signals for mesenchymal stem cell migration, proliferation and subsequently differentiation into cartilage and bone during embryonic development. A BMP when implanted with a collagenous carrier in a rat subcutaneous site is capable of inducing new bone by mimicking the cellular events of embryonic bone formation. Based on this biological principle, BMP2 and BMP7 containing collagenous matrix as carrier have been developed as bone graft substitutes for spine fusion and long bone fractures. Here, we describe a novel autologous bone graft substitute that contains BMP6 delivered within an autologous blood coagulum as carrier and summarize the biology of osteogenic BMPs in the context of bone repair and regeneration specifically the critical role that carrier plays to support osteogenesis.
Collapse
Affiliation(s)
- T Kuber Sampath
- perForm Biologics Inc., Holliston, MA 01746, United States of America.
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
38
|
Sismanoglu S, Ercal P. Dentin-Pulp Tissue Regeneration Approaches in Dentistry: An Overview and Current Trends. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1298:79-103. [PMID: 32902726 DOI: 10.1007/5584_2020_578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Conventional treatment approaches in irreversible pulpitis and apical periodontitis include the disinfection of the pulp space followed by filling with various materials, which is commonly known as the root canal treatment. Disadvantages including the loss of tooth vitality and defense mechanism against carious lesions, susceptibility to fractures, discoloration and microleakage led to the development of regenerative therapies for the dentin pulp-complex. The goal of dentin-pulp tissue regeneration is to reestablish the physiological pulp function such as pulp sensibility, pulp repair capability by mineralization and pulp immunity. Recent dentin-pulp tissue regeneration approaches can be divided into cell homing and cell transplantation. Cell based approaches include a suitable scaffold for the delivery of potent stem cells with or without bioactive molecules into the root canal system while cell homing is based on the recruitment of host endogenous stem cells from the resident tissue including periapical region or dental pulp. This review discusses the recent treatment modalities in dentin-pulp tissue regeneration through tissue engineering and current challenges and trends in this field of research.
Collapse
Affiliation(s)
- Soner Sismanoglu
- Department of Restorative Dentistry, Faculty of Dentistry, Altinbas University, Istanbul, Turkey
| | - Pınar Ercal
- Department of Oral Surgery, Faculty of Dentistry, Altinbas University, Istanbul, Turkey.
| |
Collapse
|
39
|
Sampath TK, Reddi AH. Discovery of bone morphogenetic proteins - A historical perspective. Bone 2020; 140:115548. [PMID: 32730937 DOI: 10.1016/j.bone.2020.115548] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022]
Abstract
Bone morphogenetic proteins (BMPs) were purified from demineralized bone matrix by their ability to induce new bone formation in vivo. BMPs represent a large sub-family of proteins structurally related to TGF-beta and activins. Two BMP bone graft substitutes, BMP2 (InFuse®) and BMP7 (OP1®) have been developed as products for the repair of long bone non-union fractures and lumbar spinal fusion in humans. The approval of BMP2 and BMP7 based products for use in the clinic supports that the signals responsible for bone formation at ectopic sites can form a basis as therapeutics for bone repair and regeneration. This article describes a historical perspective of the discovery BMPs.
Collapse
Affiliation(s)
- T Kuber Sampath
- perForm biologics Inc., Holliston, MA, United States of America.
| | - A Hari Reddi
- Lawrence Ellison Center for Musculoskeletal Regeneration, Department of Orthopedic Surgery, School of Medicine, University of California at Davis, Sacramento, CA, United States of America
| |
Collapse
|
40
|
Ion R, Necula MG, Mazare A, Mitran V, Neacsu P, Schmuki P, Cimpean A. Drug Delivery Systems Based on Titania Nanotubes and Active Agents for Enhanced Osseointegration of Bone Implants. Curr Med Chem 2020; 27:854-902. [PMID: 31362646 DOI: 10.2174/0929867326666190726123229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 01/16/2019] [Accepted: 05/04/2019] [Indexed: 12/31/2022]
Abstract
TiO2 nanotubes (TNTs) are attractive nanostructures for localized drug delivery. Owing to their excellent biocompatibility and physicochemical properties, numerous functionalizations of TNTs have been attempted for their use as therapeutic agent delivery platforms. In this review, we discuss the current advances in the applications of TNT-based delivery systems with an emphasis on the various functionalizations of TNTs for enhancing osteogenesis at the bone-implant interface and for preventing implant-related infection. Innovation of therapies for enhancing osteogenesis still represents a critical challenge in regeneration of bone defects. The overall concept focuses on the use of osteoconductive materials in combination with the use of osteoinductive or osteopromotive factors. In this context, we highlight the strategies for improving the functionality of TNTs, using five classes of bioactive agents: growth factors (GFs), statins, plant derived molecules, inorganic therapeutic ions/nanoparticles (NPs) and antimicrobial compounds.
Collapse
Affiliation(s)
- Raluca Ion
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Mazare
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patricia Neacsu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patrik Schmuki
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
41
|
Wang H, Deng Z, Chen J, Qi X, Pang L, Lin B, Adib YTY, Miao N, Wang D, Zhang Y, Li J, Zeng X. A novel vehicle-like drug delivery 3D printing scaffold and its applications for a rat femoral bone repairing in vitro and in vivo. Int J Biol Sci 2020; 16:1821-1832. [PMID: 32398952 PMCID: PMC7211168 DOI: 10.7150/ijbs.37552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 01/07/2020] [Indexed: 12/23/2022] Open
Abstract
The high surface area ratio and special structure of mesoporous bioactive glass (MBG) endow it with excellent physical adsorption of various drugs without destroying the chemical activity. Silicate 1393 bioactive glass (1393) is famous for its fantastic biodegradability and osteogenesis. Herein, we have built a novel vehicle-like drug delivery 3D printing scaffold with multiplexed drug delivery capacity by coating MBG on the surface of 1393 (1393@MBG). Furthermore, we have applied DEX and BMP-2 on the 1393@MBG scaffold to endow it with antibacterial and osteogenic properties. Results indicated that this 1393@MBG scaffold could effectively load and controlled release BMP-2, DNA and DEX, which can be applied for orthopedic treatment. The in vitro study showed that the DEX loaded 1393@MBG exhibited excellent antibacterial ability, which was evaluated by Staphylococcus aureus (S. aureus), and the BMP-2 loaded 1393@MBG can improve the alkaline phosphatase (ALP) activity and upregulate the expression of osteogenesis-related genes (OCN and RUNX2) of human bone mesenchymal stem cells (hBMSCs). Moreover, the in vivo study further confirmed that the BMP-2 loaded 1393@MBG group showed better osteogenic capacity as compared to that of the 1393 group in a rat femoral defect. Together, these results suggested that the vehicle-like drug delivery 3D printing scaffold 1393@MBG could be a promising candidate for bone repair and relative bone disease treatment.
Collapse
Affiliation(s)
- Hui Wang
- Laboratory for Advance Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Zhengwei Deng
- Department of Orthopedics, Fengxian District Central Hospital Affiliated of Shanghai University of Medicine&Health Sciences, 279 zhouzhu road, Shanghai 220120, People's Republic of China.,Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Chen
- Department of Orthopedics, Fengxian District Central Hospital Affiliated of Shanghai University of Medicine&Health Sciences, 279 zhouzhu road, Shanghai 220120, People's Republic of China.,Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Qi
- Department of Orthopedics, Fengxian District Central Hospital Affiliated of Shanghai University of Medicine&Health Sciences, 279 zhouzhu road, Shanghai 220120, People's Republic of China
| | - Libing Pang
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Bocai Lin
- Laboratory for Advance Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yan Teik Yuin Adib
- Laboratory for Advance Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,School of Life Science & Chemical Technology, Ngee Ann Polytechnic, Singapore 599489
| | - Na Miao
- Department of Pediatrics, Maternal and Child Health Hospital of Zaozhuang City, Shandong, China
| | - Deping Wang
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Yadong Zhang
- Department of Orthopedics, Fengxian District Central Hospital Affiliated of Shanghai University of Medicine&Health Sciences, 279 zhouzhu road, Shanghai 220120, People's Republic of China.,Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiusheng Li
- Laboratory for Advance Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xiangqiong Zeng
- Laboratory for Advance Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
42
|
Zhang B, Yang L, Zeng Z, Feng Y, Wang X, Wu X, Luo H, Zhang J, Zhang M, Pakvasa M, Wagstaff W, He F, Mao Y, Qin K, Ding H, Zhang Y, Niu C, Wu M, Zhao X, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Athiviraham A, Moriatis Wolf J, Lee MJ, Hynes K, Strelzow J, El Dafrawy M, Xia Y, He TC. Leptin Potentiates BMP9-Induced Osteogenic Differentiation of Mesenchymal Stem Cells Through the Activation of JAK/STAT Signaling. Stem Cells Dev 2020; 29:498-510. [PMID: 32041483 DOI: 10.1089/scd.2019.0292] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors that have the ability to differentiate into multiple lineages, including bone, cartilage, and fat. We previously demonstrated that the least known bone morphogenetic protein (BMP)9 (also known as growth differentiation factor 2) is one of the potent osteogenic factors that can induce both osteogenic and adipogenic differentiation of MSCs. Nonetheless, the molecular mechanism underlying BMP9 action remains to be fully understood. Leptin is an adipocyte-derived hormone in direct proportion to the amount of body fat, and exerts pleiotropic functions, such as regulating energy metabolism, bone mass, and mineral density. In this study, we investigate the potential effect of leptin signaling on BMP9-induced osteogenic differentiation of MSCs. We found that exogenous leptin potentiated BMP9-induced osteogenic differentiation of MSCs both in vitro and in vivo, while inhibiting BMP9-induced adipogenic differentiation. BMP9 was shown to induce the expression of leptin and leptin receptor in MSCs, while exogenous leptin upregulated BMP9 expression in less differentiated MSCs. Mechanistically, we demonstrated that a blockade of JAK signaling effectively blunted leptin-potentiated osteogenic differentiation induced by BMP9. Taken together, our results strongly suggest that leptin may potentiate BMP9-induced osteogenesis by cross-regulating BMP9 signaling through the JAK/STAT signaling pathway in MSCs. Thus, it is conceivable that a combined use of BMP9 and leptin may be explored as a novel approach to enhancing efficacious bone regeneration and fracture healing.
Collapse
Affiliation(s)
- Bo Zhang
- Departments of Orthopaedic Surgery and Obstetrics and Gynecology, Institute of Bone and Joint Research, The First and Second Hospitals of Lanzhou University, Lanzhou, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Lijuan Yang
- Departments of Orthopaedic Surgery and Obstetrics and Gynecology, Institute of Bone and Joint Research, The First and Second Hospitals of Lanzhou University, Lanzhou, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Breast Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaoxing Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Breast Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Breast Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Fang He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Breast Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kevin Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Huimin Ding
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Orthopaedic Surgery, BenQ Medical Center Affiliated with Nanjing Medical University, Nanjing, China
| | - Yongtao Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Laboratory Diagnostic Medicine, Chongqing General Hospital, Chongqing, China
| | - Meng Wu
- Departments of Orthopaedic Surgery and Obstetrics and Gynecology, Institute of Bone and Joint Research, The First and Second Hospitals of Lanzhou University, Lanzhou, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Xia Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Breast Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dayao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois.,Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Yayi Xia
- Departments of Orthopaedic Surgery and Obstetrics and Gynecology, Institute of Bone and Joint Research, The First and Second Hospitals of Lanzhou University, Lanzhou, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois
| |
Collapse
|
43
|
Zhang Z, Liu J, Zeng Z, Fan J, Huang S, Zhang L, Zhang B, Wang X, Feng Y, Ye Z, Zhao L, Cao D, Yang L, Pakvasa M, Liu B, Wagstaff W, Wu X, Luo H, Zhang J, Zhang M, He F, Mao Y, Ding H, Zhang Y, Niu C, Haydon RC, Luu HH, Lee MJ, Wolf JM, Shao Z, He TC. lncRNA Rmst acts as an important mediator of BMP9-induced osteogenic differentiation of mesenchymal stem cells (MSCs) by antagonizing Notch-targeting microRNAs. Aging (Albany NY) 2019; 11:12476-12496. [PMID: 31825894 PMCID: PMC6949095 DOI: 10.18632/aging.102583] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023]
Abstract
Understanding the bone and musculoskeletal system is essential to maintain the health and quality of life of our aging society. Mesenchymal stem cells (MSCs) can undergo self-renewal and differentiate into multiple tissue types including bone. We demonstrated that BMP9 is the most potent osteogenic factors although molecular mechanism underlying BMP9 action is not fully understood. Long noncoding RNAs (lncRNAs) play important regulatory roles in many physiological and/or pathologic processes. Here, we investigated the role of lncRNA Rmst in BMP9-induced osteogenic differentiation of MSCs. We found that Rmst was induced by BMP9 through Smad signaling in MSCs. Rmst knockdown diminished BMP9-induced osteogenic, chondrogenic and adipogenic differentiation in vitro, and attenuated BMP9-induced ectopic bone formation. Silencing Rmst decreased the expression of Notch receptors and ligands. Bioinformatic analysis predicted Rmst could directly bind to eight Notch-targeting miRNAs, six of which were downregulated by BMP9. Silencing Rmst restored the expression of four microRNAs (miRNAs). Furthermore, an activating Notch mutant NICD1 effectively rescued the decreased ALP activity caused by Rmst silencing. Collectively, our results strongly suggest that the Rmst-miRNA-Notch regulatory axis may play an important role in mediating BMP9-induced osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Zhicai Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jianxiang Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Shifeng Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Bo Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province, and the Departments of Orthopaedic Surgery and Obstetrics and Gynecology, The First and Second Hospitals of Lanzhou University, Lanzhou 730030, China
| | - Xi Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Zhenyu Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ling Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Daigui Cao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
- Departments of Orthopaedic Surgery and Laboratory Medicine, Chongqing General Hospital, Chongqing 400013, China
| | - Lijuan Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province, and the Departments of Orthopaedic Surgery and Obstetrics and Gynecology, The First and Second Hospitals of Lanzhou University, Lanzhou 730030, China
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bin Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xiaoxing Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jing Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Fang He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and the School of Laboratory Medicine; and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Huimin Ding
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, BenQ Medical Center Affiliated with Nanjing Medical University, Nanjing 210000, China
| | - Yongtao Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266061, China
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery and Laboratory Medicine, Chongqing General Hospital, Chongqing 400013, China
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
44
|
Weber FE. Reconsidering Osteoconduction in the Era of Additive Manufacturing. TISSUE ENGINEERING. PART B, REVIEWS 2019; 25:375-386. [PMID: 30997857 PMCID: PMC6784493 DOI: 10.1089/ten.teb.2019.0047] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023]
Abstract
Bone regeneration procedures in clinics and bone tissue engineering stand on three pillars: osteoconduction, osteoinduction, and stem cells. In the last two decades, the focus in this field has been on osteoinduction, which is realized by the use of bone morphogenetic proteins and the application of mesenchymal stem cells to treat bone defects. However, osteoconduction was reduced to a surface phenomenon because the supposedly ideal pore size of osteoconductive scaffolds was identified in the 1990s as 0.3-0.5 mm in diameter, forcing bone formation to occur predominantly on the surface. Meanwhile, additive manufacturing has evolved as a new tool to realize designed microarchitectures in bone substitutes, thereby enabling us to study osteoconduction as a true three-dimensional phenomenon. Moreover, by additive manufacturing, wide-open porous scaffolds can be produced in which bone formation occurs distant to the surface at a superior bony defect-bridging rate enabled by highly osteoconductive pores 1.2 mm in diameter. This review provides a historical overview and an updated definition of osteoconduction and related terms. In addition, it shows how additive manufacturing can be instrumental in studying and optimizing osteoconduction of bone substitutes, and provides novel optimized features and boundaries of osteoconductive microarchitectures. Impact Statement This review updates the definition of osteoconduction and draws clear lines to discriminate between osteoconduction, osseointegration, and osteoinduction. Moreover, additively manufactured libraries of scaffolds revealed that: osteoconduction is more a three-dimensional than a surface phenomenon; microarchitecture dictates defect bridging, which is the measure for osteoconduction; pore diameter or the diagonal of lattice microarchitectures of osteoconductive bone substitutes should be ∼1.2 mm.
Collapse
Affiliation(s)
- Franz E. Weber
- Oral Biotechnology and Bioengineering, Center of Dental Medicine Department of Cranio-Maxillofacial and Oral Surgery, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Abstract
OBJECTIVE Identify the molecular mechanism of inflammatory stimuli induced pancreatic cancer progression. METHODS RNA-seq, microarray assay and bioinformatics analyses were used to identify differentially expressed genes. Immunohistochemical staining was performed to evaluate CD68, CD163, β-catenin, CD103, CCL3 markers. Quantitative real-time polymerase chain reaction (qRT-PCR), luciferase reporter assay, apoptosis assay, wound healing assay and immunofluorescence were performed to study the relationship of inflammatory stimuli and WNT/β-catenin pathway. RESULTS Differentially expressed genes of macrophage-conditioned medium-treated pancreatic cancer cells were related with WNT/β-catenin pathway. Inflammatory stimuli could activate WNT/β-catenin signaling pathway. In 106 pancreatic cancer patients, nuclear β-catenin expression of CD68-high group was much higher than CD68-low group (P < 0.05), as same as CD163 (P < 0.05). Inflammatory stimuli downregulated the expression of CCL3 via WNT/β-catenin pathway and inhibited the chemotaxis of CD103 dendritic cells. Six pancreatic cancer prognosis associating genes were upregulated by inflammatory stimuli via WNT/β-catenin pathway. Transforming growth factor-β promoted malignant biological behavior of pancreatic cancer cells through WNT/β-catenin pathway-dependent mechanism. CONCLUSIONS Our present study provided a novel mechanism involved in the inflammation-driven cancer progression through tumor immune escape and downstream gene regulation of WNT/β-catenin pathway-dependent manner.
Collapse
|
46
|
Koike T, Sha J, Bai Y, Matsuda Y, Hideshima K, Yamada T, Kanno T. Efficacy of Bacterial Cellulose as a Carrier of BMP-2 for Bone Regeneration in a Rabbit Frontal Sinus Model. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2489. [PMID: 31390730 PMCID: PMC6696112 DOI: 10.3390/ma12152489] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 01/11/2023]
Abstract
If the alveolar bone height of patients requiring dental implants in the maxillary molar region is inadequate, it is difficult to achieve satisfactory outcomes using existing bone graft materials. We previously reported the possible utility of bacterial cellulose (BC) as a new dental treatment material. BC has a high absorptive capacity, good mechanical strength, and good volume retention. BC loaded with bone morphogenetic protein-2 (BMP-2) might allow effective alveolar bone augmentation. We created critical frontal bone defect models in 12 male Japanese white rabbits and divided them into four groups: sham; BC (BC grafting only); BMP-2 (treated with BMP-2 solution only); and BC+BMP-2 (grafted with BC loaded with BMP-2). Newly formed bone volume was calculated via hematoxylin-eosin staining evaluation. The proliferating cell nuclear antigen and osteocalcin levels were determined by the immunohistochemical staining analysis. All measured indices of the BC+BMP-2 group were significantly superior to those of the other groups (all p < 0.05). BC maintained the graft space and released BMP-2 in a sustained manner, promoting optimal bone formation. The BC+BMP-2 combination enhanced bone regeneration and shows promise as a useful means of clinical pre-dental implant bone augmentation in the maxillary sinus.
Collapse
Affiliation(s)
- Takashi Koike
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan
| | - Jingjing Sha
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan
| | - Yunpeng Bai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan
| | - Yuhei Matsuda
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan
| | - Katsumi Hideshima
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan
| | - Takaya Yamada
- Depart of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research, Shimane University, Izumo, Shimane 693-8501, Japan
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, 89-1 Enya-Cho, Izumo, Shimane 693-8501, Japan.
| |
Collapse
|
47
|
Maepa M, Ssemakalu CC, Motaung KS. The Potential Chondrogenic Effect of Eucomis autumnalis Aqueous Extracts on Porcine Adipose-Derived Mesenchymal Stem Cells. Tissue Eng Part A 2019; 25:1137-1145. [DOI: 10.1089/ten.tea.2018.0247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Makwese Maepa
- Department of Biomedical Sciences, Faculty of Science, Tshwane University of Technology, Pretoria, South Africa
| | - Cornelius Cano Ssemakalu
- Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark, South Africa
| | - Keolebogile Shirley Motaung
- Department of Biomedical Sciences, Faculty of Science, Tshwane University of Technology, Pretoria, South Africa
| |
Collapse
|
48
|
Liao J, Wu S, Li K, Fan Y, Dunne N, Li X. Peptide-modified bone repair materials: Factors influencing osteogenic activity. J Biomed Mater Res A 2019; 107:1491-1512. [PMID: 30790423 DOI: 10.1002/jbm.a.36663] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/29/2019] [Accepted: 02/14/2019] [Indexed: 11/08/2022]
Abstract
Many factors have been demonstrated as having an influencing effect on the osteogenic activity of the peptide-modified bone repair materials. However, most of studies only focus on one or two aspects that result in an incomplete direction for materials preparation, characterization, and performance evaluation. In this review, we reported several factors through summarizing previous research studies, which are mainly centered on three aspects: (1) the characteristics of peptide immobilized on the surface of matrix (e.g., type and length of sequence, structure, and density); (2) the combination mode between peptide and matrix (including covalent binding in selective or nonselective immobilization, and noncovalent binding through simple absorption or mixing with the matrix, and other factors in covalent binding); and (3) the properties of the matrix (including surface structure and morphology, dimension, mechanical properties, hydrophobic-hydrophilic balance, adsorbing proteins on materials), and the other possible influencing factors such as binding to other peptides. In addition, attentions were paid to the introduction and the discussion of newest studies and the analysis of mechanism. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A, 2019.
Collapse
Affiliation(s)
- Jie Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Shuai Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Kun Li
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | - Nicholas Dunne
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| |
Collapse
|
49
|
Tao J, Han Q, Zhou H, Diao X. Transcriptomic responses of regenerating earthworms (Eisenia foetida) to retinoic acid reveals the role of pluripotency genes. CHEMOSPHERE 2019; 226:47-59. [PMID: 30913427 DOI: 10.1016/j.chemosphere.2019.03.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/16/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Abstract
Exogenous retinoic acid (RA) delays and disturbs the regeneration of Eisenia foetida and inhibits the expression of pluripotent gene Sox2. However, studies of E. foetida conducted at the molecular level have been unable to elucidate its regeneration and mechanisms of RA effects on its regeneration. We merged existing transcriptomic data for E. foetida to generate a high-confidence set of transcriptomes. The de novo assembly of transcriptomes was performed by using the Trinity method, and functional annotations were analysed. We performed RNA-seq on four samples of regenerating tail fragments, three across a time-course (0, 3 and 7 days post amputation) and the fourth sample exposed to RA (7 days post amputation). E. foetida regeneration genes underwent significant upregulation and downregulation over the examined time periods, which may have been caused by a shared regulatory programme controlled by multiple gene families. The inhibition of RA against earthworm regeneration is likely related to the expression of these genes. Using annotation data and clustering, we also identified specific transcripts of 6 gene superfamilies enriched among genes exhibiting differential expression during regeneration periods and exhibiting the same expression patterns as those of the Sox2 gene. The regeneration transcriptome of tail fragment regeneration serves as a strong resource for investigating global expression changes that occur during regeneration and the toxicity of RA. This study offers insight for better understanding the regeneration of lower animals and molecular mechanisms of RA toxicity in invertebrates.
Collapse
Affiliation(s)
- Jing Tao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; College of Life Sciences and Pharmacy, Hainan University, Haikou, 570228, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Qian Han
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; College of Life Sciences and Pharmacy, Hainan University, Haikou, 570228, China.
| | - Hailong Zhou
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; College of Life Sciences and Pharmacy, Hainan University, Haikou, 570228, China.
| | - Xiaoping Diao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China; College of Life Science, Hainan Normal University, Haikou, 571158, China.
| |
Collapse
|
50
|
Um IW, Ku JK, Lee BK, Yun PY, Lee JK, Nam JH. Postulated release profile of recombinant human bone morphogenetic protein-2 (rhBMP-2) from demineralized dentin matrix. J Korean Assoc Oral Maxillofac Surg 2019; 45:123-128. [PMID: 31334099 PMCID: PMC6620303 DOI: 10.5125/jkaoms.2019.45.3.123] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Demineralized dentin matrix (DDM) has been used as a recombinant human bone morphogenetic protein-2 (rhBMP-2) carrier in many clinical trials. To optimize the clinical safety and efficacy of rhBMP-2 with DDM, efforts have been made to improve the delivery of rhBMP-2 by 1) lowering the administered dose, 2) localizing the protein, and 3) prolonging its retention time at the action site as well as the bone forming capacity of the carrier itself. The release profile of rhBMP-2 that is associated with endogenous BMP in dentin has been postulated according to the type of incorporation, which is attributed to the loosened interfibrillar space and nanoporous dentinal tubule pores. Physically adsorbed and modified, physically entrapped rhBMP-2 is sequentially released from the DDM surface during the early stage of implantation. As DDM degradation progresses, the loosened interfibrillar space and enlarged dentinal tubules release the entrapped rhBMP-2. Finally, the endogenous BMP in dentin is released with osteoclastic dentin resorption. According to the postulated release profile, DDM can therefore be used in a controlled manner as a sequential delivery scaffold for rhBMP-2, thus sustaining the rhBMP-2 concentration for a prolonged period due to localization. In addition, we attempted to determine how to lower the rhBMP-2 concentration to 0.2 mg/mL, which is lower than the approved 1.5 mg/mL.
Collapse
Affiliation(s)
- In-Woong Um
- R&D Institute, Korea Tooth Bank, Seoul, Korea
| | - Jeong-Kui Ku
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Armed Forces Capital Hospital, Seongnam, Korea.,Department of Oral and Maxillofacial Surgery, Seoul Asan Medical Center, Seoul, Korea
| | - Bu Kyu Lee
- Department of Oral and Maxillofacial Surgery, Seoul Asan Medical Center, Seoul, Korea
| | - Pil-Young Yun
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jeong Keun Lee
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Science, Ajou University School of Medicine, Suwon, Korea
| | - Jeong-Hun Nam
- Department of Dental Implant/Oral Surgery, Private Clinic, Seoul, Korea
| |
Collapse
|