1
|
Chen Q, Liu S, Wang Y, Tong M, Sun H, Dong M, Lu Y, Niu W, Wang L. Yam Carbon Dots Promote Bone Defect Repair by Modulating Histone Demethylase 4B. Int J Nanomedicine 2024; 19:10415-10434. [PMID: 39430312 PMCID: PMC11491100 DOI: 10.2147/ijn.s477587] [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: 05/09/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Chronic apical periodontitis is a typical inflammatory disease of the oral cavity, the pathology is characterized by an inflammatory reaction with bone defects in the periapical area. Chinese medicine is our traditional medicine, Carbon Dots (CDs) are a new type of nanomaterials. The purpose of this study was to prepare Yam Carbon Dots (YAM-CDs) to investigate the mechanism of action of YAM-CDs on bone differentiation in vivo and in vitro. Methods We characterized YAM-CDs using transmission electron microscopy (TEM), Fourier Transform Infrared Spectrometer (FTIR), X-Ray Diffraction (XRD) and photoluminescence (PL). CCK-8 assay, Real-time qPCR, and Western Blot were conducted using bone marrow mesenchymal stem cells (BMSCs) to verify that YAM-CDs promote osteoblast differentiation. In addition, we investigated the role of YAM-CDs in promoting bone formation in an inflammatory setting in an in vivo mouse model of cranial defects. Results The results of TEM and PL showed that the YAM-CDs mostly consisted of the components C1s, O1s, and N1s. Additionally the average sizes of YAM-CDs were 2-6 nm. The quantum yield was 4.44%, with good fluorescence stability and biosafety. Real-time qPCR and Western blot analysis showed that YAM-CDs promoted osteoblast differentiation under an inflammatory environment by regulating expression of histone demethylase 4B (KDM4B). In vivo, results showed that YAM-CDs effectively repaired cranial bone defects in a mouse model and reduced the expression of inflammatory factors under the action of lipopolysaccharides (LPS). Conclusion YAM-CDs promoted the proliferation and differentiation of osteoblasts by regulating the expression of KDM4B to repair cranial bone defects in mice under an LPS-induced inflammatory milieu, which will provide a new idea for the treatment of clinical periapical inflammation and other bone defect diseases.
Collapse
Affiliation(s)
- QianYang Chen
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - Shuo Liu
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - Yuhan Wang
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - MeiChen Tong
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - HaiBo Sun
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - Ming Dong
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - Yun Lu
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - WeiDong Niu
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| | - LiNa Wang
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
| |
Collapse
|
2
|
Kubi JA, Brah AS, Cheung KMC, Lee YL, Lee KF, Sze SCW, Qiao W, Yeung KWK. A new osteogenic protein isolated from Dioscorea opposita Thunb accelerates bone defect healing through the mTOR signaling axis. Bioact Mater 2023; 27:429-446. [PMID: 37152710 PMCID: PMC10160600 DOI: 10.1016/j.bioactmat.2023.04.018] [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] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023] Open
Abstract
Delayed bone defect repairs lead to severe health and socioeconomic impacts on patients. Hence, there are increasing demands for medical interventions to promote bone defect healing. Recombinant proteins such as BMP-2 have been recognized as one of the powerful osteogenic substances that promote mesenchymal stem cells (MSCs) to osteoblast differentiation and are widely applied clinically for bone defect repairs. However, recent reports show that BMP-2 treatment has been associated with clinical adverse side effects such as ectopic bone formation, osteolysis and stimulation of inflammation. Here, we have identified one new osteogenic protein, named 'HKUOT-S2' protein, from Dioscorea opposita Thunb. Using the bone defect model, we have shown that the HKUOT-S2 protein can accelerate bone defect repair by activating the mTOR signaling axis of MSCs-derived osteoblasts and increasing osteoblastic biomineralization. The HKUOT-S2 protein can also modulate the transcriptomic changes of macrophages, stem cells, and osteoblasts, thereby enhancing the crosstalk between the polarized macrophages and MSCs-osteoblast differentiation to facilitate osteogenesis. Furthermore, this protein had no toxic effects in vivo. We have also identified HKUOT-S2 peptide sequence TKSSLPGQTK as a functional osteogenic unit that can promote osteoblast differentiation in vitro. The HKUOT-S2 protein with robust osteogenic activity could be a potential alternative osteoanabolic agent for promoting osteogenesis and bone defect repairs. We believe that the HKUOT-S2 protein may potentially be applied clinically as a new class of osteogenic agent for bone defect healing.
Collapse
Affiliation(s)
- John Akrofi Kubi
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Augustine Suurinobah Brah
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Kenneth Man Chee Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, HKU, 21 Sassoon Road, Hong Kong S.A.R, PR China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, HKU- Shenzhen Hospital, Shenzhen, PR China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, HKU, 21 Sassoon Road, Hong Kong S.A.R, PR China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, HKU- Shenzhen Hospital, Shenzhen, PR China
| | - Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong S.A.R, PR China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong S.A.R, PR China
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Hong Kong S.A.R, PR China
| | - Kelvin Wai-Kwok Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| |
Collapse
|
3
|
Lee JK, Ha JH, Kim DK, Kwon J, Cho YE, Kwun IS. Depletion of Zinc Causes Osteoblast Apoptosis with Elevation of Leptin Secretion and Phosphorylation of JAK2/STAT3. Nutrients 2022; 15:nu15010077. [PMID: 36615735 PMCID: PMC9824754 DOI: 10.3390/nu15010077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Zinc (Zn) has been reported to mediate leptin secretion, and thus leptin can be an important candidate molecule linking Zn with bone formation. The present study investigated whether zinc deficiency induces leptin secretion by activating a JAK2/STAT3 signaling pathway and leads to osteoblastic apoptosis. MC3T3-E1 cells were incubated for 24 h in normal osteogenic differentiation medium (OSM) or OSM treated with either 1 μM (Low Zn) or 15 μM (High Zn) of ZnCl2 containing 5 μM TPEN (Zn chelator). Our results demonstrated that low Zn stimulated extracellular leptin secretion and increased mRNA and protein expression of leptin in osteoblastic MC3T3-E1 cells. The OB-Rb (long isoform of leptin receptor) expressions were also elevated in osteoblasts under depletion of Zn. Leptin-signaling proteins, JAK2 and p-JAK2 in the cytosol of low Zn osteoblast conveyed leptin signaling, which ultimately induced higher p-STAT3 expression in the nucleus. Apoptotic effects of JAK2/STAT3 pathway were shown by increased caspase-3 in low Zn osteoblasts as well as apoptotic morphological features observed by TEM. Together, these data suggest that low Zn modulates leptin secretion by activating JAK2/STAT3 signaling pathway and induces apoptosis of osteoblastic MC3T3-E1 cells.
Collapse
Affiliation(s)
- Jennifer K. Lee
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Republic of Korea
| | - Do-Kyun Kim
- Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - JaeHee Kwon
- Department of Food and Nutrition, Andong National University, 388 Songchundong, Andong 36729, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, 388 Songchundong, Andong 36729, Republic of Korea
- Correspondence: (Y.-E.C.); (I.-S.K.); Tel.: +82-54-820-5484 (Y.-E.C.)
| | - In-Sook Kwun
- Department of Food and Nutrition, Andong National University, 388 Songchundong, Andong 36729, Republic of Korea
- Correspondence: (Y.-E.C.); (I.-S.K.); Tel.: +82-54-820-5484 (Y.-E.C.)
| |
Collapse
|
4
|
Ge Y, Ding S, Feng J, Du J, Gu Z. Diosgenin inhibits Wnt/β-catenin pathway to regulate the proliferation and differentiation of MG-63 cells. Cytotechnology 2021; 73:169-178. [PMID: 33927474 DOI: 10.1007/s10616-021-00454-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/17/2021] [Indexed: 12/18/2022] Open
Abstract
Dioscorea opposita Thunb has the effect of anti-osteoporosis, but whether its active ingredient diosgenin (DIO) has an anti-osteoporosis effect is unknown. The purpose of this study is to investigate the effect of DIO on the proliferation and differentiation of MG-63 cells. MG-63 cells were treated with different concentrations of DIO (0.001, 0.01, 0.1 and 1 μM) or 20 mM Wnt/β-catenin signaling agonist-LiCl, and then their cell cycle and viability were analyzed by flow cytometry and 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT), respectively. To investigate osteoblast differentiation, alizarin red staining and ultraviolet spectrophotometer were used to determine the number of calcified nodules and the activity of alkaline phosphatase (ALP), respectively. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting were used to detect the expressions of proliferation-related, osteogenic-related and Wnt/β-catenin signal pathway-related factors. After the cells were treated with low-concentration (0.001 or 0.01 μM) DIO, cell viability was significantly increased and the proportion of cells in S phase was increased. In addition, low-concentration DIO could significantly increase the expression of Ki67, proliferating cell nuclear antigen (PCNA), osteopontin (OPN), and osteocalcin (BGP), promote osteoblast differentiation, and suppress the expression of β-catenin, Runx2 and cyclinD1. However, high concentrations of DIO showed the opposite effect. Low-concentration DIO obviously reversed the effect of LiCl on decreasing the number of calcified nodules and inhibiting the expression of OPN and BGP in cells. Low-concentration DIO might promote the proliferation and differentiation of MG-63 cell by inhibiting the Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Yunlin Ge
- The Fourth Department of Orthopedics, The 903th Hospital of People's Liberation Army, Hangzhou, China
| | - Shuchen Ding
- The Fifth Department of Orthopedics, The 903th Hospital of People's Liberation Army, Hangzhou, China
| | - Jiangbiao Feng
- The Fourth Department of Orthopedics, The 903th Hospital of People's Liberation Army, Hangzhou, China
| | - Jiangdong Du
- The Fourth Department of Orthopedics, The 903th Hospital of People's Liberation Army, Hangzhou, China
| | - Zenghui Gu
- The Third Department of Orthopedics, The 903th Hospital of People's Liberation Army, No.40 Jichang Road, Jianggan District, Hangzhou, 310001 Zhejiang Province China
| |
Collapse
|
5
|
Gardin C, Bosco G, Ferroni L, Quartesan S, Rizzato A, Tatullo M, Zavan B. Hyperbaric Oxygen Therapy Improves the Osteogenic and Vasculogenic Properties of Mesenchymal Stem Cells in the Presence of Inflammation In Vitro. Int J Mol Sci 2020; 21:ijms21041452. [PMID: 32093391 PMCID: PMC7073059 DOI: 10.3390/ijms21041452] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperbaric oxygen (HBO) therapy has been reported to be beneficial for treating many conditions of inflammation-associated bone loss. The aim of this work was to in vitro investigate the effect of HBO in the course of osteogenesis of human Mesenchymal Stem Cells (MSCs) grown in a simulated pro-inflammatory environment. Cells were cultured with osteogenic differentiation factors in the presence or not of the pro-inflammatory cytokine Tumor Necrosis Factor-α (TNF-α), and simultaneously exposed daily for 60 min, and up to 21 days, at 2,4 atmosphere absolute (ATA) and 100% O2. To elucidate osteogenic differentiation-dependent effects, cells were additionally pre-committed prior to treatments. Cell metabolic activity was evaluated by means of the MTT assay and DNA content quantification, whereas osteogenic and vasculogenic differentiation was assessed by quantification of extracellular calcium deposition and gene expression analysis. Metabolic activity and osteogenic properties of cells did not differ between HBO, high pressure (HB) alone, or high oxygen (HO) alone and control if cells were pre-differentiated to the osteogenic lineage. In contrast, when treatments started contextually to the osteogenic differentiation of the cells, a significant reduction in cell metabolic activity first, and in mineral deposition at later time points, were observed in the HBO-treated group. Interestingly, TNF-α supplementation determined a significant improvement in the osteogenic capacity of cells subjected to HBO, which was not observed in TNF-α-treated cells exposed to HB or HO alone. This study suggests that exposure of osteogenic-differentiating MSCs to HBO under in vitro simulated inflammatory conditions enhances differentiation towards the osteogenic phenotype, providing evidence of the potential application of HBO in all those processes requiring bone regeneration.
Collapse
Affiliation(s)
- Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Gerardo Bosco
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Quartesan
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Alex Rizzato
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Marco Tatullo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| |
Collapse
|
6
|
Bone-forming peptide-3 induces osteogenic differentiation of bone marrow stromal cells via regulation of the ERK1/2 and Smad1/5/8 pathways. Stem Cell Res 2018; 26:28-35. [DOI: 10.1016/j.scr.2017.11.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022] Open
|
7
|
Starch-derived absorbable polysaccharide hemostat enhances bone healing via BMP-2 protein. Acta Histochem 2017; 119:257-263. [PMID: 28168995 DOI: 10.1016/j.acthis.2017.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/27/2017] [Indexed: 11/22/2022]
Abstract
Surgical hemostasis is critical in reducing the likelihood of excessive bleeding and blood transfusion. In treating some cases, commonly used hemostatic agent showed limited efficacy and prolonged degradation and clearance, causing an inhibition of bone healing. Starch absorbable polysaccharide (SAPH) is a novel hemostatic agent made from a plant starch, which can be completely absorbed and achieve better hemostatic effects than many commonly used hemostatic agents. However, whether SAPH can induce a promotion of bone healing remains unknown. In this study, we used a model of rabbit parietal bone defect and a mouse osteoblast cell line MC3T3-E1 to evaluate the effects of SAPH on bone healing. We found that SAPH significantly decreased bone healing scores, reduced defective area of parietal bone, and increased the areas of bone trabeculae and cavitas medullaris. In addition, SAPH enhanced MC3T3-E1 osteoblasts proliferation, up-regulated the expressions of alkaline phosphatase (ALP) and osteocalcin and increased the level of bone morphogenetic protein 2 (BMP-2) in MC3T3-E1 osteoblasts. These SAPH-induced benefits in MC3T3-E1 osteoblasts were significantly abolished by the application of BMP-2-siRNA. These findings suggested that SAPH enhances bone healing, promotes the proliferation, differentiation and maturation of osteoblast by up-regulating BMP-2 expression in osteoblastic cells.
Collapse
|
8
|
You M, Jing J, Tian D, Qian J, Yu G. Dioscin stimulates differentiation of mesenchymal stem cells towards hypertrophic chondrocytes in vitro and endochondral ossification in vivo. Am J Transl Res 2016; 8:3930-3938. [PMID: 27725872 PMCID: PMC5040690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/07/2016] [Indexed: 06/06/2023]
Abstract
Dioscin has been shown to play important roles in suppression of osteoclast maturation. It is proposed as a potential natural product for the treatment of osteoclast-related diseases. We hypothesized in this study that treatment of dioscin on bone marrow mesenchymal stem cells (BMSCs) could increase the osteo-chondrogenic differentiation of BMSCs and promote endochondral ossification of BMSCs in bone fracture environment. BMSCs were extracted from femur and tibia of male C57b mice. Stemness of BMSCs was studied by performing proliferation assay and multilineage differentiation. Glycosaminoglycans (GAG) and collagen contents were assessed to examine the chondrogenesis of BMSCs. Real time quantitative PCR was carried out to examine the expression of hypertrophic marker collagen type X. Efficacy of Dioscin was then tested in mouse bone fracture model on the distal side of femur. Results showed treatment of dioscin on BMSCs increased chondrogenic differentiation of BMSCs as well as the expression of collagen type X. Local delivery of dioscin promoted endochondral ossification at bone fractured site, as shown by histological examination. Results of immunohistochemistry showed that dioscin increased collagen type X expression in bone facture model of mice. In conclusion, our results demonstrated that treatment of dioscin promote the hypertrophic differentiation of BMSCs derived chondrocytes. Dioscin could be a useful drug to promote bone regeneration after fracture.
Collapse
Affiliation(s)
- Murong You
- Department of Orthopedics, Jiangxi Provincial People’s HospitalNanchang 330006, Jiangxi Province, People’s Republic of China
| | - Juehua Jing
- Department of Orthopaedics, The Second Hospital of Anhui Medical UniversityHefei 230601, Anhui Province, People’s Republic of China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Hospital of Anhui Medical UniversityHefei 230601, Anhui Province, People’s Republic of China
| | - Jun Qian
- Department of Orthopaedics, The Second Hospital of Anhui Medical UniversityHefei 230601, Anhui Province, People’s Republic of China
| | - Guangrong Yu
- Department of Orthopaedics, The Second Hospital of Anhui Medical UniversityHefei 230601, Anhui Province, People’s Republic of China
| |
Collapse
|
9
|
Che CT, Wong MS, Lam CWK. Natural Products from Chinese Medicines with Potential Benefits to Bone Health. Molecules 2016; 21:239. [PMID: 26927052 PMCID: PMC6274145 DOI: 10.3390/molecules21030239] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/03/2016] [Accepted: 02/12/2016] [Indexed: 01/23/2023] Open
Abstract
Osteoporosis is a progressive, systemic bone disorder characterized by loss of bone mass and microstructure, leading to reduced bone strength and increased risk of fracture. It is often associated with reduced quality of life and other medical complications. The disease is common in the aging population, particularly among postmenopausal women and patients who receive long-term steroidal therapy. Given the rapid growth of the aging population, increasing life expectancy, the prevalence of bone loss, and financial burden to the healthcare system and individuals, demand for new therapeutic agents and nutritional supplements for the management and promotion of bone health is pressing. With the advent of global interest in complementary and alternative medicine and natural products, Chinese medicine serves as a viable source to offer benefits for the improvement and maintenance of bone health. This review summarizes the scientific information obtained from recent literatures on the chemical ingredients of Chinese medicinal plants that have been reported to possess osteoprotective and related properties in cell-based and/or animal models. Some of these natural products (or their derivatives) may become promising leads for development into dietary supplements or therapeutic drugs.
Collapse
Affiliation(s)
- Chun-Tao Che
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Man Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Christopher Wai Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| |
Collapse
|