1
|
Ali M, Kim YS. A comprehensive review and advanced biomolecule-based therapies for osteoporosis. J Adv Res 2024:S2090-1232(24)00215-7. [PMID: 38810908 DOI: 10.1016/j.jare.2024.05.024] [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: 03/26/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND The prevalence of osteoporosis (OP) on a global scale is significantly elevated that causes life threatening issues. The potential of groundbreaking biomolecular therapeutics in the field of OP is highly encouraging. The administration of biomolecular agents has the potential to mitigate the process of bone demineralization while concurrently augmenting the regenerative capacity of bone tissue, thereby facilitating a personalized therapeutic approach. Biomolecules-based therapies showed promising results in term of bone mass protection and restoration in OP. AIM OF REVIEW We summarized the recent biomolecular therapies with notable progress in clinical, demonstrating the potential to transform illness management. These treatments frequently utilize different biomolecule based strategies. Biomolecular therapeutics has a targeted character, which results in heightened specificity and less off-target effects, ultimately leading to increased patient outcomes. These aspects have the capacity to greatly enhance the management of OP, thus resulting in a major enhancement in the quality of life encountered by individuals affected by this condition.
Collapse
Affiliation(s)
- Maqsood Ali
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, Republic of Korea
| | - Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, Republic of Korea; Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Chungnam 31151, Republic of Korea.
| |
Collapse
|
2
|
Rastogi SK, Khanka S, Kumar S, Lakra A, Rathur R, Sharma K, Bisen AC, Bhatta RS, Kumar R, Singh D, Sinha AK. Design, synthesis and biological evaluation of novel pyrimidine derivatives as bone anabolic agents promoting osteogenesis via the BMP2/SMAD1 signaling pathway. RSC Med Chem 2024; 15:677-694. [PMID: 38389884 PMCID: PMC10880903 DOI: 10.1039/d3md00500c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/16/2023] [Indexed: 02/24/2024] Open
Abstract
Anti-resorptive inhibitors such as bisphosphonates are widely used but they have limited efficacy and serious side effects. Though subcutaneous injection of teriparatide [PTH (1-34)] is an effective anabolic therapy, long-term repeated subcutaneous administration is not recommended. Henceforth, orally bio-available small-molecule-based novel therapeutics are unmet medical needs to improve the treatment. In this study, we designed, synthesized, and carried out a biological evaluation of 31 pyrimidine derivatives as potent bone anabolic agents. A series of in vitro experiments confirmed N-(5-bromo-4-(4-bromophenyl)-6-(2,4,5-trimethoxyphenyl)pyrimidin-2-yl)hexanamide (18a) as the most efficacious anabolic agent at 1 pM. It promoted osteogenesis by upregulating the expression of osteogenic genes (RUNX2 and type 1 col) via activation of the BMP2/SMAD1 signaling pathway. In vitro osteogenic potential was further validated using an in vivo fracture defect model where compound 18a promoted the bone formation rate at 5 mg kg-1. We also established the structure-activity relationship and pharmacokinetic studies of 18a.
Collapse
Affiliation(s)
- Sumit K Rastogi
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Sonu Khanka
- Division of Endocrinology, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Santosh Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Amardeep Lakra
- Division of Endocrinology, CSIR-Central Drug Research Institute Lucknow 226031 India
| | - Rajat Rathur
- Division of Endocrinology, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Kriti Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Amol Chhatrapati Bisen
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute Lucknow 226031 India
| | - Rabi Sankar Bhatta
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute Lucknow 226031 India
| | - Ravindra Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Divya Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| | - Arun K Sinha
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002. U.P. India
| |
Collapse
|
3
|
Gipson GR, Nolan K, Kattamuri C, Kenny AP, Agricola Z, Edwards NA, Zinski J, Czepnik M, Mullins MC, Zorn AM, Thompson TB. Formation and characterization of BMP2/GDF5 and BMP4/GDF5 heterodimers. BMC Biol 2023; 21:16. [PMID: 36726183 PMCID: PMC9893541 DOI: 10.1186/s12915-023-01522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Proteins of the TGFβ family, which are largely studied as homodimers, are also known to form heterodimers with biological activity distinct from their component homodimers. For instance, heterodimers of bone morphogenetic proteins, including BMP2/BMP7, BMP2/BMP6, and BMP9/BMP10, among others, have illustrated the importance of these heterodimeric proteins within the context of TGFβ signaling. RESULTS In this study, we have determined that mature GDF5 can be combined with mature BMP2 or BMP4 to form BMP2/GDF5 and BMP4/GDF5 heterodimer. Intriguingly, this combination of a BMP2 or BMP4 monomer, which exhibit high affinity to heparan sulfate characteristic to the BMP class, with a GDF5 monomer with low heparan sulfate affinity produces a heterodimer with an intermediate affinity. Using heparin affinity chromatography to purify the heterodimeric proteins, we then determined that both the BMP2/GDF5 and BMP4/GDF5 heterodimers consistently signaled potently across an array of cellular and in vivo systems, while the activities of their homodimeric counterparts were more context dependent. These differences were likely driven by an increase in the combined affinities for the type 1 receptors, Alk3 and Alk6. Furthermore, the X-ray crystal structure of BMP2/GDF5 heterodimer was determined, highlighting the formation of two asymmetric type 1 receptor binding sites that are both unique relative to the homodimers. CONCLUSIONS Ultimately, this method of heterodimer production yielded a signaling molecule with unique properties relative to the homodimeric ligands, including high affinity to multiple type 1 and moderate heparan binding affinity.
Collapse
Affiliation(s)
- Gregory R Gipson
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kristof Nolan
- Department of Biochemistry and Molecular Biophysics, University of Chicago, Chicago, IL, USA
| | - Chandramohan Kattamuri
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alan P Kenny
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zachary Agricola
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nicole A Edwards
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joseph Zinski
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Magdalena Czepnik
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron M Zorn
- Perinatal Institute, Divisions of Developmental Biology and Neonatology & Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Thomas B Thompson
- Department of Molecular & Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Hu Y, Huang J, Chen C, Wang Y, Hao Z, Chen T, Wang J, Li J. Strategies of Macrophages to Maintain Bone Homeostasis and Promote Bone Repair: A Narrative Review. J Funct Biomater 2022; 14:18. [PMID: 36662065 PMCID: PMC9864083 DOI: 10.3390/jfb14010018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Bone homeostasis (a healthy bone mass) is regulated by maintaining a delicate balance between bone resorption and bone formation. The regulation of physiological bone remodeling by a complex system that involves multiple cells in the skeleton is closely related to bone homeostasis. Loss of bone mass or repair of bone is always accompanied by changes in bone homeostasis. However, due to the complexity of bone homeostasis, we are currently unable to identify all the mechanisms that affect bone homeostasis. To date, bone macrophages have been considered a third cellular component in addition to osteogenic spectrum cells and osteoclasts. As confirmed by co-culture models or in vivo experiments, polarized or unpolarized macrophages interact with multiple components within the bone to ensure bone homeostasis. Different macrophage phenotypes are prone to resorption and formation of bone differently. This review comprehensively summarizes the mechanisms by which macrophages regulate bone homeostasis and concludes that macrophages can control bone homeostasis from osteoclasts, mesenchymal cells, osteoblasts, osteocytes, and the blood/vasculature system. The elaboration of these mechanisms in this narrative review facilitates the development of macrophage-based strategies for the treatment of bone metabolic diseases and bone defects.
Collapse
Affiliation(s)
- Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jinghuan Huang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200000, China
| | - Chunying Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| |
Collapse
|
5
|
Cefazolin/BMP-2-Loaded Mesoporous Silica Nanoparticles for the Repair of Open Fractures with Bone Defects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8385456. [PMID: 36193077 PMCID: PMC9526639 DOI: 10.1155/2022/8385456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/20/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022]
Abstract
The study aimed to explore the feasibility of a nanodrug delivery system to treat open fractures with bone defects. We developed a cefazolin (Cef)/bone morphogenetic protein 2 (BMP-2)@mesoporous silica nanoparticle (MSN) delivery system; meanwhile, Cef/MBP-2@ poly(lactic-co-glycolic acid) (PLGA) was also developed as control. For the purpose of determining the osteogenic and anti-inflammatory actions of the nanodelivery system, we cultured bone marrow mesenchymal stem cells (BMSCs) and constructed a bone defect mouse model to evaluate its clinical efficacy. After physicochemical property testing, we determined that MSN had good stability and did not easily accumulate or precipitate and it could effectively prolong the Cef’s half-life by nearly eight times. In BMSCs, we found that compared with the PLGA delivery system, MSNs better penetrated into the bone tissue, thus effectively increasing BMSCs’ proliferation and migration ability to facilitate bone defect repair. Furthermore, the MSN delivery system could improve BMSCs’ mineralization indexes (alkaline phosphatase [ALP], osteocalcin [OCN], and collagen I [Col I]) to effectively improve its osteogenic ability. Moreover, the MSN delivery system could inhibit inflammation in bone defect mice, which was mainly reflected in its ability to reduce the release of IL-1β and IL-4 and increase IL-10 levels; it could also effectively reduce apoptosis of CD4+ and CD8+ T cells, thus improving their immune function. Furthermore, the percentage of new bones, bone mineral density, trabecular volume, and trabecular numbers in the fracture region were improved in mice treated with MSN, which allowed better repair of bone defects. Hence, Cef/BMP-2@MSN may be feasible for open fractures with bone defects.
Collapse
|
6
|
Tanaka Y, Aung KT, Ono M, Mikai A, Dang AT, Hara ES, Tosa I, Ishibashi K, Ono-Kimura A, Nawachi K, Kuboki T, Oohashi T. Suppression of Bone Necrosis around Tooth Extraction Socket in a MRONJ-like Mouse Model by E-rhBMP-2 Containing Artificial Bone Graft Administration. Int J Mol Sci 2021; 22:ijms222312823. [PMID: 34884630 PMCID: PMC8657653 DOI: 10.3390/ijms222312823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 11/30/2022] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is related to impaired bone healing conditions in the maxillomandibular bone region as a complication of bisphosphonate intake. Although there are several hypotheses for the onset of MRONJ symptoms, one of the possible causes is the inhibition of bone turnover and blood supply leading to bone necrosis. The optimal treatment strategy for MRONJ has not been established either. BMP-2, a member of the TGF-β superfamily, is well known for regulating bone remodeling and homeostasis prenatally and postnatally. Therefore, the objectives of this study were to evaluate whether cyclophosphamide/zoledronate (CY/ZA) induces necrosis of the bone surrounding the tooth extraction socket, and to examine the therapeutic potential of BMP-2 in combination with the hard osteoinductive biomaterial, β-tricalcium phosphate (β-TCP), in the prevention and treatment of alveolar bone loss around the tooth extraction socket in MRONJ-like mice models. First, CY/ZA was intraperitoneally administered for three weeks, and alveolar bone necrosis was evaluated before and after tooth extraction. Next, the effect of BMP-2/β-TCP was investigated in both MRONJ-like prevention and treatment models. In the prevention model, CY/ZA was continuously administered for four weeks after BMP-2/β-TCP transplantation. In the treatment model, CY/ZA administration was suspended after transplantation of BMP-2/β-TCP. The results showed that CY/ZA induced a significant decrease in the number of empty lacunae, a sign of bone necrosis, in the alveolar bone around the tooth extraction socket after tooth extraction. Histological analysis showed a significant decrease in the necrotic alveolar bone around tooth extraction sockets in the BMP-2/β-TCP transplantation group compared to the non-transplanted control group in both MRONJ-like prevention and treatment models. However, bone mineral density, determined by micro-CT analysis, was significantly higher in the BMP-2/β-TCP transplanted group than in the control group in the prevention model only. These results clarified that alveolar bone necrosis around tooth extraction sockets can be induced after surgical intervention under CY/ZA administration. In addition, transplantation of BMP-2/β-TCP reduced the necrotic alveolar bone around the tooth extraction socket. Therefore, a combination of BMP-2/β-TCP could be an alternative approach for both prevention and treatment of MRONJ-like symptoms.
Collapse
Affiliation(s)
- Yukie Tanaka
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Y.T.); (A.T.D.); (K.I.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
| | - Kyaw Thu Aung
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Y.T.); (A.T.D.); (K.I.); (T.O.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan; (A.M.); (A.O.-K.); (K.N.)
- Correspondence: ; Tel.: +81-86-235-7129; Fax: +81-86-222-7768
| | - Akihiro Mikai
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan; (A.M.); (A.O.-K.); (K.N.)
| | - Anh Tuan Dang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Y.T.); (A.T.D.); (K.I.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
| | - Emilio Satoshi Hara
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Ikue Tosa
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
| | - Kei Ishibashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Y.T.); (A.T.D.); (K.I.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
| | - Aya Ono-Kimura
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan; (A.M.); (A.O.-K.); (K.N.)
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kumiko Nawachi
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan; (A.M.); (A.O.-K.); (K.N.)
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (K.T.A.); (I.T.); (T.K.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan; (A.M.); (A.O.-K.); (K.N.)
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Y.T.); (A.T.D.); (K.I.); (T.O.)
| |
Collapse
|
7
|
Choi LY, Kim MH, Yang WM. Promotion of osteogenesis by Sweroside via BMP2-involved signaling in postmenopausal osteoporosis. Phytother Res 2021; 35:7050-7063. [PMID: 34818696 DOI: 10.1002/ptr.7336] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 01/09/2023]
Abstract
Phlomis umbrosa has been traditionally used for bone diseases in traditional Korean Medicine. Sweroside (SOS), marker compounds of P. umbrosa, has been known to promote osteoblast differentiation. In this study, ameliorative effects of SOS on osteoporosis and potential target pathway were investigated. Ovariectomized mice were administered three doses of SOS three times a week for 4 weeks after inducing osteoporosis. Bone mineral content (BMC) and bone mineral density (BMD) were analyzed by dual energy X-ray absorptiometry. A human osteosarcoma cell line (SaOS-2) was differentiated to clarify the promoting effects of SOS on osteoblast differentiation and bone formation. Osteoblastic bone-forming markers were evaluated in lumbar vertebrae (LV) and mineralized SaOS-2 cells. SOS markedly elevated BMC and BMD levels and attenuated the bone marrow adipocytes in the femoral shaft. SOS increased the formation of bone matrix in SaOS-2 cells. Bone morphogenetic protein-2 (BMP2) and runt-related transcription factor 2 (CBFA1) in LV and SaOS-2 cells were up-regulated by SOS. SOS increased alkaline phosphatase (ALPL), osteopontin (SPP1), and bone sialoprotein-1 (BSPH1). In conclusion, SOS induced the formation of mineralized bone matrix by regulating BMP2/CBFA1-mediated molecules. Therefore, SOS could be a therapeutic compound of treatment for osteoporosis by producing the new bone matrix.
Collapse
Affiliation(s)
- La Yoon Choi
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Mi Hye Kim
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Kim JH, Kim M, Hong S, Kim EY, Lee H, Jung HS, Sohn Y. Albiflorin Promotes Osteoblast Differentiation and Healing of Rat Femoral Fractures Through Enhancing BMP-2/Smad and Wnt/β-Catenin Signaling. Front Pharmacol 2021; 12:690113. [PMID: 34349649 PMCID: PMC8327266 DOI: 10.3389/fphar.2021.690113] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Fracture healing is related to osteogenic differentiation and mineralization. Recently, due to the unwanted side effects and clinical limitations of existing treatments, various natural product-based chemical studies have been actively conducted. Albiflorin is a major ingredient in Paeonia lactiflora, and this study investigated its ability to promote osteogenic differentiation and fracture healing. To demonstrate the effects of albiflorin on osteoblast differentiation and calcified nodules, alizarin red S staining and von Kossa staining were used in MC3T3-E1 cells. In addition, BMP-2/Smad and Wnt/β-catenin mechanisms known as osteoblast differentiation mechanisms were analyzed through RT-PCR and western blot. To investigate the effects of albiflorin on fracture healing, fractures were induced using a chainsaw in the femur of Sprague Dawley rats, and then albiflorin was intraperitoneally administered. After 1, 2, and 3 weeks, bone microstructure was analyzed using micro-CT. In addition, histological analysis was performed by staining the fractured tissue, and the expression of osteogenic markers in serum was measured. The results demonstrated that albiflorin promoted osteoblastogenesis and the expression of RUNX2 by activating BMP-2/Smad and Wnt/β-catenin signaling in MC3T3-E1 cells. In addition, albiflorin upregulated the expression of various osteogenic genes, such as alkaline phosphatase, OCN, bone sialoprotein, OPN, and OSN. In the femur fracture model, micro-CT analysis showed that albiflorin played a positive role in the formation of callus in the early stage of fracture recovery, and histological examination proved to induce the expression of osteogenic genes in femur tissue. In addition, the expression of bone-related genes in serum was also increased. This suggests that albiflorin promotes osteogenesis, bone calcification and bone formation, thereby promoting the healing of fractures in rats.
Collapse
Affiliation(s)
- Jae-Hyun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Minsun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - SooYeon Hong
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun-Young Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyangsook Lee
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
9
|
Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: Pre-clinical studies. Bone 2021; 143:115758. [PMID: 33212318 PMCID: PMC7769985 DOI: 10.1016/j.bone.2020.115758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022]
Abstract
During the past several years, pre-clinical experiments have established that microRNAs (miRNAs), small non-coding RNAs, serve as key regulatory molecules of fracture healing. Their easy modulation with agonists and antagonists make them highly desirable targets for future therapeutic strategies, especially for pathophysiologic fractures that either do not heal (nonunions) or are delayed. It is now well documented that these problematic fractures lead to human suffering and impairment of life quality. Additionally, financial difficulties are also encountered as work productivity decreases and income is reduced. Moreover, targeting miRNAs may also be an avenue to enhancing normal physiological fracture healing. Herein we present the most current knowledge of the involvement of miRNAs during fracture healing in pre-clinical studies. Following a brief description on the nature of miRNAs and of the fracture healing process, we present data from studies focusing specifically, on miRNA regulation of osteoblast differentiation and osteogenesis (within the context of known signaling pathways), chondrocytes, angiogenesis, and apoptosis, all critical to successful bone repair. Further, we also discuss miRNAs and exosomes. We hope that this manuscript serves as a comprehensive review that will facilitate basic/translational scientists in the orthopaedic arena to realize and further decipher the biological and future therapeutic impact of these small regulatory RNA molecules, especially as they relate to the molecular events of each of the major phases of fracture healing.
Collapse
Affiliation(s)
- David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Edie Duque
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, United States of America.
| |
Collapse
|
10
|
Shockwave Therapy Modulates the Expression of BMP2 for Prevention of Bone and Cartilage Loss in the Lower Limbs of Postmenopausal Osteoporosis Rat Model. Biomedicines 2020; 8:biomedicines8120614. [PMID: 33333838 PMCID: PMC7765335 DOI: 10.3390/biomedicines8120614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 01/25/2023] Open
Abstract
Osteoporosis (OP) causes bone loss and weakness, increasing the risk of bone fracture. In this study, rats were divided into Sham, OP, SW(F) (0.25 mJ/mm2 with 1600 impulses to the left medial femur), and SW(T) (0.25 mJ/mm2 with 1600 impulses to the left medial tibia). The bone strength results following SW(T) were better than SW(F) in the modulus, extension at peak load, handleability, and strain at break. SW(T) had the best prevention for bone loss in both lower limbs of ovariectomized (OVX) rats. The cartilage cellular matrixes of both knees were improved in SW(T) and SW(F) compared to that of OP. Serum bone morphogenetic protein 2 (BMP2) in rats undergoing SW(T) or SW(F) was significantly improved compared to that in Sham and OP. The expressions of BMP2, BMP4, and SMAD family member 4 (Smad4) in addition to the Wnt family member 3A (Wnt3a) and Cyclin D1 signaling key factors were significantly induced in the cartilage of both knees by shockwave (SW). SW(T) presented the best efficacy to induce serum BMP2 to prevent bone loss from both lower limbs. Here, we display the protective effects of SW therapy to induce BMP2, BMP4, Smad4, Wnt3a, and Cyclin D1 signaling factors for cartilage loss in both knees of OVX rats.
Collapse
|
11
|
Nirmala FS, Lee H, Kim JS, Ha T, Jung CH, Ahn J. Green Tomato Extract Prevents Bone Loss in Ovariectomized Rats, a Model of Osteoporosis. Nutrients 2020; 12:nu12103210. [PMID: 33096661 PMCID: PMC7589907 DOI: 10.3390/nu12103210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/14/2020] [Indexed: 01/04/2023] Open
Abstract
Although drug therapies are available for postmenopausal osteoporosis, these drugs are not free of side effects and long-term adherence to them are low. A safe and effective nutritional approach to counter postmenopausal osteoporosis is an important research goal. We fed ovariectomized (OVX) Sprague–Dawley rats a diet supplemented with 1% or 2% green tomato extract (GTE). After 12 weeks, micro-computed tomography scans revealed that GTE supplementation effectively prevented distal femur bone loss. This prevention was due to improved bone formation and suppressed bone resorption as observed by the regulation of osteoblast and osteoclast activities. GTE supplementation also improved bone formation through Bmp2-Smad 1/5/8-Runx2 signaling, while bone resorption was regulated by the receptor activator of nuclear factor kappa-B (RANKL)/osteoprogeterin (OPG) pathway. These results suggest that GTE supplementation prevents severe postmenopausal bone loss by maintaining the regulation of bone homeostasis in OVX rats. GTE as a diet supplement might be a potential novel alternative for the prevention of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Farida S. Nirmala
- Department of Food Biotechnology, University of Science and Technology, Daejeon 305350, Korea; (F.S.N.); (T.H.); (C.H.J.)
| | - Hyunjung Lee
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju 55365, Korea; (H.L.); (J.-S.K.)
| | - Ji-Sun Kim
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju 55365, Korea; (H.L.); (J.-S.K.)
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea
| | - Taeyoul Ha
- Department of Food Biotechnology, University of Science and Technology, Daejeon 305350, Korea; (F.S.N.); (T.H.); (C.H.J.)
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju 55365, Korea; (H.L.); (J.-S.K.)
| | - Chang Hwa Jung
- Department of Food Biotechnology, University of Science and Technology, Daejeon 305350, Korea; (F.S.N.); (T.H.); (C.H.J.)
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju 55365, Korea; (H.L.); (J.-S.K.)
| | - Jiyun Ahn
- Department of Food Biotechnology, University of Science and Technology, Daejeon 305350, Korea; (F.S.N.); (T.H.); (C.H.J.)
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju 55365, Korea; (H.L.); (J.-S.K.)
- Correspondence:
| |
Collapse
|
12
|
Muñoz J, Akhavan NS, Mullins AP, Arjmandi BH. Macrophage Polarization and Osteoporosis: A Review. Nutrients 2020; 12:nu12102999. [PMID: 33007863 PMCID: PMC7601854 DOI: 10.3390/nu12102999] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Over 200 million people suffer from osteoporosis worldwide. Individuals with osteoporosis have increased rates of bone resorption while simultaneously having impaired osteogenesis. Most current treatments for osteoporosis focus on anti-resorptive methods to prevent further bone loss. However, it is important to identify safe and cost-efficient treatments that not only inhibit bone resorption, but also stimulate anabolic mechanisms to upregulate osteogenesis. Recent data suggest that macrophage polarization may contribute to osteoblast differentiation and increased osteogenesis as well as bone mineralization. Macrophages exist in two major polarization states, classically activated macrophages (M1) and alternatively activated macrophage (M2) macrophages. The polarization state of macrophages is dependent on molecules in the microenvironment including several cytokines and chemokines. Mechanistically, M2 macrophages secrete osteogenic factors that stimulate the differentiation and activation of pre-osteoblastic cells, such as mesenchymal stem cells (MSC’s), and subsequently increase bone mineralization. In this review, we cover the mechanisms by which M2 macrophages contribute to osteogenesis and postulate the hypothesis that regulating macrophage polarization states may be a potential treatment for the treatment of osteoporosis.
Collapse
|
13
|
Durbano HW, Halloran D, Nguyen J, Stone V, McTague S, Eskander M, Nohe A. Aberrant BMP2 Signaling in Patients Diagnosed with Osteoporosis. Int J Mol Sci 2020; 21:ijms21186909. [PMID: 32967078 PMCID: PMC7555210 DOI: 10.3390/ijms21186909] [Citation(s) in RCA: 14] [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: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
The most common bone disease in humans is osteoporosis (OP). Current therapeutics targeting OP have several negative side effects. Bone morphogenetic protein 2 (BMP2) is a potent growth factor that is known to activate both osteoblasts and osteoclasts. It completes these actions through both SMAD-dependent and SMAD-independent signaling. A novel interaction between the BMP type Ia receptor (BMPRIa) and casein kinase II (CK2) was discovered, and several CK2 phosphorylation sites were identified. A corresponding blocking peptide (named CK2.3) was designed to further elucidate the phosphorylation site’s function. Previously, CK2.3 demonstrated an increased osteoblast activity and decreased osteoclast activity in a variety of animal models, cell lines, and isolated human osteoblasts. It is hypothesized that CK2.3 completes these actions through the BMP signaling pathway. Furthermore, it was recently discovered that BMP2 did not elicit an osteogenic response in osteoblasts from patients diagnosed with OP, while CK2.3 did. In this study, we explore where in the BMP pathway the signaling disparity or defect lies in those diagnosed with OP. We found that osteoblasts isolated from patients diagnosed with OP did not activate SMAD or ERK signaling after BMP2 stimulation. When OP osteoblasts were stimulated with BMP2, both BMPRIa and CK2 expression significantly decreased. This indicates a major disparity within the BMP signaling pathway in patients diagnosed with osteoporosis.
Collapse
Affiliation(s)
- Hilary W. Durbano
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (H.W.D.); (D.H.); (J.N.); (V.S.)
| | - Daniel Halloran
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (H.W.D.); (D.H.); (J.N.); (V.S.)
| | - John Nguyen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (H.W.D.); (D.H.); (J.N.); (V.S.)
| | - Victoria Stone
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (H.W.D.); (D.H.); (J.N.); (V.S.)
| | - Sean McTague
- Christiana Care Hospital, Newark, DE 19716, USA; (S.M.); (M.E.)
| | - Mark Eskander
- Christiana Care Hospital, Newark, DE 19716, USA; (S.M.); (M.E.)
| | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (H.W.D.); (D.H.); (J.N.); (V.S.)
- Correspondence: ; Tel.: +1-302-831-2959
| |
Collapse
|
14
|
Hendrijantini N, Hartono CK, Daniati RP, Hong G, Sitalaksmi RM, Kuntjoro M, Ari MDA. Human Umbilical Cord Mesenchymal Stem Cell-induced Osterix, Bone Morphogenetic Protein-2, and Tartrate-resistant Acid Phosphatase Expression in Osteoporotic Mandibular Bone. Eur J Dent 2020; 15:84-89. [PMID: 32898872 PMCID: PMC7902117 DOI: 10.1055/s-0040-1715987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVES The aim of this study was to prove that human umbilical cord mesenchymal stem cell (hUCMSC) therapy conducted according to the mandibular osteoporotic model will increase Osterix (Osx) and bone morphogenetic protein-2 (BMP-2) expression, while reducing tartrate-resistant acid phosphatase (TRAP) expression. PKH26 labeling proves that mandibular bone regeneration is produced by hUCMSCs induction. MATERIALS AND METHODS This study incorporated a true posttest only control group design. Twenty-five female Wistar rats were randomly divided into five groups consisting of the sham surgery (N) group, osteoporotic groups injected with gelatin for 4 weeks (G4) and 8 weeks (G8), and osteoporotic groups injected with hUCMSC-gelatin for 4weeks (SC4) and 8 weeks (SC8). All subjects were provided for BMP-2, Osx, and TRAP on immunohistochemistry examination and PKH-26 labeling. STATISTICAL ANALYSIS All data were analyzed using ANOVA and Tukey HSD tests with p < 0.05 being considered as statistically significant. RESULTS Compared with other groups, the highest level of BMP-2 and Osx occurred in the sham surgery (N) and osteoporotic groups injected with hUCMSCs-gelatin (SC), while the lowest level of TRAP was found in SC4. During 4- and 8-week observation periods, the PKH 26 appeared green (fluorescent). CONCLUSIONS hUCMSC demonstrates high-osteogenic activity and increased osteoporotic mandibular bone regeneration, as shown by increased expression of Osx and BMP-2 and decreased TRAP expression. From the labeling, PKH-26 proved that viable hUCMSCs in gelatin solvent can be present in the mandibular bone and be capable of promoting osteogenic differentiation and increasing mineralization and bone formation in the osteoporotic mandibular bone.
Collapse
Affiliation(s)
- Nike Hendrijantini
- Department of Prosthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Cindy Karina Hartono
- Department of Prosthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Reni Puspa Daniati
- Department of Prosthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Guang Hong
- Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Aoba-ku, Sendai, Japan
| | - Ratri Maya Sitalaksmi
- Department of Prosthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Mefina Kuntjoro
- Department of Prosthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | |
Collapse
|
15
|
A Synthetic Peptide, CK2.3, Inhibits RANKL-Induced Osteoclastogenesis through BMPRIa and ERK Signaling Pathway. J Dev Biol 2020; 8:jdb8030012. [PMID: 32660129 PMCID: PMC7557985 DOI: 10.3390/jdb8030012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 12/23/2022] Open
Abstract
The skeletal system plays an important role in the development and maturation process. Through the bone remodeling process, 10% of the skeletal system is renewed every year. Osteoblasts and osteoclasts are two major bone cells that are involved in the development of the skeletal system, and their activity is kept in balance. An imbalance between their activities can lead to diseases such as osteoporosis that are characterized by significant bone loss due to the overactivity of bone-resorbing osteoclasts. Our laboratory has developed a novel peptide, CK2.3, which works as both an anabolic and anti-resorptive agent to induce bone formation and prevent bone loss. We previously reported that CK2.3 mediated mineralization and osteoblast development through the SMAD, ERK, and AKT signaling pathways. In this study, we demonstrated the mechanism by which CK2.3 inhibits osteoclast development. We showed that the inhibition of MEK by the U0126 inhibitor rescued the osteoclast development of RAW264.7 induced by RANKL in a co-culture system with CK2.3. We observed that CK2.3 induced ERK activation and BMPRIa expression on Day 1 after stimulation with CK2.3. While CK2.3 was previously reported to induce the SMAD signaling pathway in osteoblast development, we did not observe any changes in SMAD activation in osteoclast development with CK2.3 stimulation. Understanding the mechanism by which CK2.3 inhibits osteoclast development will allow CK2.3 to be developed as a new treatment for osteoporosis.
Collapse
|
16
|
Xie H, Liu M, Jin Y, Lin H, Zhang Y, Zheng S. miR-1323 suppresses bone mesenchymal stromal cell osteogenesis and fracture healing via inhibiting BMP4/SMAD4 signaling. J Orthop Surg Res 2020; 15:237. [PMID: 32600409 PMCID: PMC7322887 DOI: 10.1186/s13018-020-01685-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Atrophic non-union fractures show no radiological evidence of callus formation within 3 months of fracture. microRNA dysregulation may underlie the dysfunctional osteogenesis in atrophic non-union fractures. Here, we aimed to analyze miR-1323 expression in human atrophic non-union fractures and examine miR-1323’s underlying mechanism of action in human mesenchymal stromal cells. Methods Human atrophic non-union and standard healing fracture specimens were examined using H&E and Alcian Blue staining, immunohistochemistry, qRT-PCR, immunoblotting, and ALP activity assays. The effects of miR-1323 mimics or inhibition on BMP4, SMAD4, osteogenesis-related proteins, ALP activity, and bone mineralization were analyzed in human mesenchymal stromal cells. Luciferase reporter assays were utilized to assay miR-1323’s binding to the 3'UTRs of BMP4 and SMAD4. The effects of miR-1323, BMP4, and SMAD4 were analyzed by siRNA and overexpression vectors. A rat femur fracture model was established to analyze the in vivo effects of antagomiR-1323 treatment. Results miR-1323 was upregulated in human atrophic non-union fractures. Atrophic non-union was associated with downregulation of BMP4 and SMAD4 as well as the osteogenic markers ALP, collagen I, and RUNX2. In vitro, miR-1323 suppressed BMP4 and SMAD4 expression by binding to the 3'UTRs of BMP4 and SMAD4. Moreover, miR-1323’s inhibition of BMP4 and SMAD4 inhibited mesenchymal stromal cell osteogenic differentiation via modulating the nuclear translocation of the transcriptional co-activator TAZ. In vivo, antagomiR-1323 therapy facilitated the healing of fractures in a rat model of femoral fracture. Conclusions This evidence supports the miR-1323/BMP4 and miR-1323/SMAD4 axes as novel therapeutic targets for atrophic non-union fractures.
Collapse
Affiliation(s)
- Hui Xie
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Ming Liu
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yaofeng Jin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Haiqing Lin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yushan Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Song Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China.
| |
Collapse
|
17
|
Chen J, Liu M, Luo X, Peng L, Zhao Z, He C, He Y. Exosomal miRNA-486-5p derived from rheumatoid arthritis fibroblast-like synoviocytes induces osteoblast differentiation through the Tob1/BMP/Smad pathway. Biomater Sci 2020; 8:3430-3442. [PMID: 32406432 DOI: 10.1039/c9bm01761e] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of rheumatoid arthritis (RA) is related to the inhibition of osteoblast differentiation. Exosomes secreted from RA fibroblast-like synoviocytes (RA-FLSs-exos) are associated with the pathogenesis of RA and microRNAs (miRNAs) being crucial for RA progression. Accordingly, the aim of the present study is to elucidate the effect of RA-FLS-derived exosomes on osteoblast differentiation and further identify exosomal cargos responsible for this effect. RA-FLSs were isolated from a RA patient and osteoblasts from the donor bone. Isolated RA-FLSs-exos were co-cultured with osteoblasts. Osteoblast differentiation was evaluated by ALP quantification assays, Alizarin Red S staining, and determining markers of osteoblast activity (Osx, OC, Col1a1 and Dlx2). Collagen induced arthritis (CIA)-induced mouse models were established. RA-FLSs-exo could be phagocytosed by osteoblasts. Elevating the expression of miR-486-5p in RA-FLSs-exo promoted osteoblast differentiation. miR-486-5p targeted Tob1 and activated the BMP/Smad signaling pathway in osteoblasts. In addition, RA-FLSs-exo containing miR-486-5p facilitated osteoblast differentiation by activating the BMP/Smad signaling pathway and repressing Tob1. Moreover, RA-FLSs-exo containing miR-486-5p alleviated the disease severity of RA by decreasing Tob1 expression in CIA-induced mice. To sum up, RA-FLSs-exo carrying miR-486-5p serve as a promoter for osteoblast differentiation in RA, ultimately highlighting a promising competitive new target for RA treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
18
|
Abourehab MAS. Hyaluronic Acid Modified Risedronate and Teriparatide Co-loaded Nanocarriers for Improved Osteogenic Differentiation of Osteoblasts for the Treatment of Osteoporosis. Curr Pharm Des 2020; 25:2975-2988. [PMID: 31368869 DOI: 10.2174/1381612825666190801140703] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Owing to its multifactorial intricate pathogenesis, combined therapeutic regimen is considered appropriate for the treatment of osteoporosis. However, a multi-drug regimen is also associated with adverse effects due to the non-specific distribution of drugs. Therefore, the present study aims for efficient codelivery of risedronate (RDN) (a potent bone anti-resorptive drug) and teriparatide (TPD) (anabolic agent) as hyaluronic acid (HA)-modified chitosan nanoparticles (NPs). METHODS RDN/TPD NPs were synthesized using the high- pressure homogenization - solvent evaporation technique. The fabricated NPs were then characterized and optimized for suitable physicochemical characteristics. The optimized NPs were then evaluated for bone remodeling potential via assessment of time-mannered modulation in proliferation, differentiation, and mineralization of osteoblasts. RESULTS Results showed that HA-RDN/TPD NPs exhibited excellent physicochemical characteristics (nanoscopic size, stable zeta potential, high entrapment efficiency, and smooth spherical shape) and remained stable upon storage in the refrigerator. Assessment of various aspects of the cell growth cycle (i.e., proliferation, differentiation, and mineralization) evidenced promising bone regeneration efficacy of HA-RDN/TPD NPs. CONCLUSION This new strategy of employing simultaneous delivery of anti-resorptive and bone-forming agents would open new horizons for scientists, researchers, and healthcare providers as an efficient pharmacotherapy for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt.,Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
19
|
Zhang T, Wei Q, Fan D, Liu X, Li W, Song C, Tian Y, Cai H, Zheng Y, Liu Z. Improved osseointegration with rhBMP-2 intraoperatively loaded in a specifically designed 3D-printed porous Ti6Al4V vertebral implant. Biomater Sci 2019; 8:1279-1289. [PMID: 31867583 DOI: 10.1039/c9bm01655d] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Three-dimensional (3D)-printed porous Ti6Al4V implants are commonly used for reconstructing bone defects in the treatment of orthopaedic diseases owing to their excellent osteoconduction. However, to achieve improved therapeutic outcomes, the osteoinduction of these implants requires further improvement. The aim of this study was to investigate the combined use of recombinant human BMP-2 (rhBMP-2) with a 3D-printed artificial vertebral implant (3D-AVI) to improve the osteoinduction. Eight male Small Tail Han sheep underwent cervical corpectomy, and 3D-AVIs with or without loaded rhBMP-2 in cavities designed at the center were implanted to treat the cervical defect. Radiographic, micro-computed tomography, fluorescence labelling, and histological examination revealed that the osseointegration efficiency of the rhBMP-2 group was significantly higher than that of the blank control group. The biomechanical test results suggested that rhBMP-2 reduced the range of motion of the cervical spine and provided a more stable implant. Fluorescence observations revealed that the bone tissue grew from the periphery to the center of the 3D-AVIs, first growing into the pore space and then interlocking with the Ti6Al4V implant surface. Therefore, we successfully improved osseointegration of the 3D-AVI by loading rhBMP-2 into the cavity designed at the center of the Ti6Al4V implant, realizing earlier and more stable fixation of implants postoperatively in a simple manner. These benefits of rhBMP-2 are expected to expand the application range and reliability of 3D-printed porous Ti6Al4V implants and improve their therapeutic efficacy.
Collapse
Affiliation(s)
- Teng Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Chang B, Qiu H, Zhao H, Yang X, Wang Y, Ji T, Zhang Y, Quan Q, Li Y, Zeng J, Meng H, Gu Y. The Effects of Photobiomodulation on MC3T3-E1 Cells via 630 nm and 810 nm Light-Emitting Diode. Med Sci Monit 2019; 25:8744-8752. [PMID: 31743330 PMCID: PMC6880645 DOI: 10.12659/msm.920396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Photobiomodulation (PBM) has been explored as a promising therapeutic strategy to regulate bone cell growth; however, the effects of PBM on osteoblast cell lines remains poorly understood. In addition, as a light source of PBM, the light uniformity of light-emitting diode (LED) devices has not been given enough attention. MATERIAL AND METHODS Here, we sought to investigate the effects of PBM on MC3T3-E1 cells via 630 nm and 810 nm light from a newly designed LED with high uniformity of light. Cell proliferation, flow cytometric analysis, alkaline phosphatase (ALP) staining, ALP activity, Alizarin Red S staining, and quantitative real-time polymerase chain reaction (qRT-PCR) were carried out to assess treatment response. MC3T3-E1 cells were irradiated with LED devices (630±5 nm and 810±10 nm, continuous wave) for 200 seconds at a power density of 5 mW/cm² once daily. RESULTS Increases in cell proliferation and decreases in cell apoptosis were evident following irradiation. ALP staining intensity and activity were also significantly increased following irradiation. Level of mineralization was obviously enhanced in irradiated groups compared with non-irradiated controls. qRT-PCR also showed significant increases in mRNA expression of osteocalcin (OCN) and osteoprotegerin (OPG) in the irradiated groups. CONCLUSIONS Our results showed that LED PBM could promote the proliferation, ALP staining intensity and activity, level of mineralization, gene expression of OCN and OPG of MC3T3-E1 cells, with no significant difference between the 630 nm- and 810 nm-irradiated groups.
Collapse
Affiliation(s)
- Biao Chang
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Haixia Qiu
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Hongyou Zhao
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Xi Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, P.R. China
- General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, P.R. China
| | - Ying Wang
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Tengda Ji
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Yuxuan Zhang
- Institute of Orthopedics, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, P.R. China
- Key Laboratory of Musculoskeletal Trauma and War Injuries, People’s Liberation Army, Beijing, P.R. China
| | - Qi Quan
- Institute of Orthopedics, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, P.R. China
- Key Laboratory of Musculoskeletal Trauma and War Injuries, People’s Liberation Army, Beijing, P.R. China
| | - Yunqi Li
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Jing Zeng
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Haoye Meng
- Institute of Orthopedics, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, P.R. China
- Key Laboratory of Musculoskeletal Trauma and War Injuries, People’s Liberation Army, Beijing, P.R. China
| | - Ying Gu
- Department of Laser Medicine, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| |
Collapse
|
21
|
Mei F, Tu Y. Cyclophilin B enhances the proliferation and differentiation of MC3T3-E1 cells via JAK2/STAT3 signaling pathway. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1684842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Fan Mei
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, Hubei, PR China
| | - Yanhong Tu
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, Hubei, PR China
| |
Collapse
|
22
|
Guo W, Shi K, Xiang G, Lu D, Dou H, Xie C, Chen L. Effects of Rhizoma Drynariae Cataplasm on Fracture Healing in a Rat Model of Osteoporosis. Med Sci Monit 2019; 25:3133-3139. [PMID: 31030207 PMCID: PMC6503747 DOI: 10.12659/msm.914568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Osteoporosis is an increasingly prevalent disease characterized by decreased bone mass and deterioration of the bone microstructure, which contribute to increased fragility and subsequent fragility fractures, especially in elderly individuals. Rhizoma Drynariae (DRE) is among the most frequently used herbal medicines for the treatment of osteoporosis. Transdermal delivery is a proven novel pathway for drug treatment and has several advantages over traditional drug delivery routes. MATERIAL AND METHODS Female Sprague-Dawley osteoporotic fracture model rats were divided into 3 groups: the control group, the DRE (90 mg/kg/day) group and the DRE cataplasm (containing 30 mg DRE, administered at right femur site daily) group. At 3 and 6 weeks after operation, we performed x-ray, histological, and biomechanical analyses, and evaluated bone marrow density of the femur. RESULTS Treatment with DRE increased callus formation and bone union compared with the control group. Moreover, DRE enhanced bone strength at the femoral diaphysis in the osteoporotic fractures in rats by increasing the ultimate load and stiffness compared with the control group. Furthermore, DRE restored the trabecular bone mineral density in the femur compared with the control group. DRE cataplasm application further enhanced the therapeutic effects against osteoporotic fracture in this rat model. CONCLUSIONS DRE cataplasm application might be useful against osteoporotic fracture.
Collapse
Affiliation(s)
- Weijun Guo
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Kesi Shi
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Guangheng Xiang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Di Lu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Haicheng Dou
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Chenglong Xie
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Long Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| |
Collapse
|
23
|
Zhu W, Ding W, Shang X, Zhu D, Dai X. Fangchinoline Promotes Autophagy and Inhibits Apoptosis in Osteoporotic Rats. Med Sci Monit 2019; 25:324-332. [PMID: 30632520 PMCID: PMC6338015 DOI: 10.12659/msm.912624] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Osteoporosis is a common disorder leading to bone loss. At present, the treatment options available for the management of osteoporosis are limited. The present investigation evaluated the protective effect of fangchinoline against osteoporosis and also postulates the possible mechanism of action. MATERIAL AND METHODS Osteoporosis was induced by subcutaneously injecting prednisolone (2.5 mg/pellet) for 4 weeks. Fangchinoline 1, 3 and 10 mg/kg was given intraperitoneally for the period. Protective effects of fangchinoline were assessed by estimating microarchitectural parameters and bone mineral density (BMD) in the vertebrae tissues, and biochemical parameters were also determined in the serum of rats with prednisolone-induced osteoporosis. Moreover, gene expression of microtubule-associated protein 1A/1B-light chain 3 (LC3), B cell lymphoma 2 (Bcl-2), caspase-3, bone morphogenetic protein 2 (BMP2), Beclin-1, autophagy-related 5 (ATG-5), Runt-related transcription factor 2 (RUNX-2), and receptor activator of nuclear factor kappa-b ligand (RANKL) protein in the vertebrae tissue were assessed by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot assay. RESULTS There was a significant (p<0.01) decrease in the BMD and microarchitectural parameters in the vertebrae tissue of the fangchinoline-treated group compared to the prednisolone group. We also found that treatment with fangchinoline attenuated the altered expressions of LC3, Bcl-2, caspase-3, BMP2, Beclin-1, ATG-5, RUNX-2, and RANKL protein in the prednisolone-induced osteoporosis rats. Moreover, levels of biochemical parameters were attenuated in the serum of fangchinoline-treated and prednisolone-induced osteoporosis rat. Histopathology revealed that the apoptosis of osteoblasts was decreased in the fangchinoline-treated group compared to the prednisolone group of rats. CONCLUSIONS Fangchinoline inhibits apoptosis of osteoblasts and protects against bone loss in prednisolone-induced osteoporosis rats by inducing autophagy.
Collapse
|