1
|
Shen J, Hu C, Wang Y, Tan Y, Gao X, Zhang N, Lv J, Sun J. The SRC/NF-κB-AKT/NOS3 axis as a key mediator of Kaempferol's protective effects against oxidative stress-induced osteoclastogenesis. Immun Inflamm Dis 2024; 12:e70045. [PMID: 39422344 PMCID: PMC11488077 DOI: 10.1002/iid3.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Osteoclasts are integral to the advancement of osteoporosis (OP), and their generation under conditions of oxidative stress (OS) involves various pathways. However, the specific mechanism through which the natural antioxidant kaempferol (KAE) mitigates the influence of OS on osteoclasts remains somewhat uncertain. This study aims to evaluate the effect of KAE on osteoclast formation under OS and explore its possible mechanism. METHODS Zebrafish were used to observe the effects of KAE on OP and OS. OP and OS "double disease targets" network pharmacology were used to predict the action target and mechanism of KAE on OP under OS. The effects of KAE on osteoclast differentiation induced by OS were evaluated using RWA264.7 cells induced by LPS. To elucidate the potential mechanism, we detected the expression of related factors and target genes during induction. RESULTS The presence of KAE exhibited potential in improving the conditions of OP and OS in zebrafish. KAE can reduce the OS of RAW 264.7 cells stimulated by LPS, inhibit the formation of osteoclasts, and change the level of related factors of OS, and reduce the increase of TRAP. The utilization of network pharmacology and target gene expression assay revealed that KAE exerted a down-regulatory effect on the expression of proto-oncogene tyrosine protein kinase (SRC), nuclear factor kappa-B (NF-κB), Serine/Threonine Kinase-1 (AKT1), Nitric Oxide Synthase 3 (NOS3) and Matrix Metallopeptidase-2 (MMP2). CONCLUSION Based on the results of this study, KAE may effectively mitigate OS and impede the formation of osteoclasts through the SRC/NF-κB-AKT/NOS3 axis.
Collapse
Affiliation(s)
- Jiaming Shen
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Chunjie Hu
- Affiliated HospitalChangchun University of Chinese MedicineChangchunChina
| | - Yuelong Wang
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Yiying Tan
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Xiaochen Gao
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Nanxi Zhang
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Jingwei Lv
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Jiaming Sun
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| |
Collapse
|
2
|
Zhou YS, Huang J, Cao WX, Yu AX, Li P, Liang JL, Leng XY, Jin J, Yu P, Liu J. The therapeutic mechanism of Compound Lurong Jiangu Capsule for the treatment of cadmium-induced osteoporosis: network pharmacology and experimental verification. Front Endocrinol (Lausanne) 2024; 15:1331488. [PMID: 39050570 PMCID: PMC11266182 DOI: 10.3389/fendo.2024.1331488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/03/2024] [Indexed: 07/27/2024] Open
Abstract
Background Among bone diseases, osteoporosis-like skeleton, such as trabecular thinning, fracture and so on, is the main pathological change of cadmium-induced osteoporosis(Cd-OP), accompanied by brittle bone and increased fracture rate. However, the mechanism underlying cadmium-induced osteoporosis has remained elusive. Compound Lurong Jiangu Capsule (CLJC) is an experienced formula for the treatment of bone diseases, which has the effect of tonifying kidney and strengthening bones, promoting blood circulation and relieving pain. Objective Network pharmacology and molecular docking technology combined with experiments were used to investigate the potential mechanism of CLJC in treating Cd-OP. Method The active compounds and corresponding targets of each herb in CLJC were searched in the TCMSP and BATMAN-TCM databases. The DisGeNet, OMIM, and GeneCards databases searched for Cd-OP targets. The relationship between both of them was visualized by establishing an herb-compound-target network using Cytoscape 3.9.1 software. Gene ontology (GO), and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were performed after determining the intersection of the targets from CLJC and Cd-OP. What's more, molecular docking was performed to validate the results. All of them were aim to obtain hud signaling pathways for further study. Finally, BAX, BCL-2, and CASPASE-3 were screened and selected for further experiments, which included bone imaging and reconstruction analysis (Micro-CT), hematoxylin-eosin Staining (HE), and western blot (WB). Results 106 common targets from CLJC and Cd-OP targets were identified. KEGG pathway analysis suggested that multiple signaling pathways, such as the pathways in cancer, may play roles in treatment. Verification of the molecular docking was successful. Here we showed that Cd-OP displayed Tb.Th and Tb.N significantly reduced and even broke, irregular proliferation of bone cortex, uneven and loose trabecular bone arrangement, changed in apoptosis-related proteins, such as significant upregulation of CASPASE-3, BAX protein and significant downregulation of BCL-2 protein in vivo, while CLJC rescued these phenotypes. Conclusion This study revealed that CLJC can reduce the expression of apoptosis-related proteins, and multiple components and multiple targets inhibit Cd-OP through apoptosis signaling pathway.
Collapse
Affiliation(s)
- Ya-shuang Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jian Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, China
| | - Wen-xuan Cao
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ao-xue Yu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Pan Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jin-ling Liang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiang-yang Leng
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jian Jin
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Yu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jia Liu
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
3
|
Yu X, Ren Z, Wang Y, Yuan G, Hu J, Song L, Pan C, Feng K, Liu Y, Shao L, Zhang L, Wang J, Zhao J, Bao N, Sun Z. Kaempferol attenuates particle-induced osteogenic impairment by regulating ER stress via the IRE1α-XBP1s pathway. J Biol Chem 2024; 300:107394. [PMID: 38768813 PMCID: PMC11223082 DOI: 10.1016/j.jbc.2024.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Periprosthetic osteolysis and subsequent aseptic loosening are the primary causes of failure following total joint arthroplasty. Wear particle-induced osteogenic impairment is recognized as an important contributing factor in the development of osteolysis, with endoplasmic reticulum (ER) stress emerging as a pivotal underlying mechanism. Hence, searching for potential therapeutic targets and agents capable of modulating ER stress in osteoblasts is crucial for preventing aseptic loosening. Kaempferol (KAE), a natural flavonol compound, has shown promising osteoprotective effects and anti-ER stress properties in diverse diseases. However, the influence of KAE on ER stress-mediated osteogenic impairment induced by wear particles remains unclear. In this study, we observed that KAE effectively relieved TiAl6V4 particles-induced osteolysis by improving osteogenesis in a mouse calvarial model. Furthermore, we demonstrated that KAE could attenuate ER stress-mediated apoptosis in osteoblasts exposed to TiAl6V4 particles, both in vitro and in vivo. Mechanistically, our results revealed that KAE mitigated ER stress-mediated apoptosis by upregulating the IRE1α-XBP1s pathway while concurrently partially inhibiting the IRE1α-regulated RIDD and JNK activation. Collectively, our findings suggest that KAE is a prospective therapeutic agent for treating wear particle-induced osteolysis and highlight the IRE1α-XBP1s pathway as a potential therapeutic target for preventing aseptic loosening.
Collapse
Affiliation(s)
- Xin Yu
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yuxiang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Guodong Yuan
- Department of Orthopedics, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianlun Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Cheng Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kangkang Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yuqiao Liu
- Medical Information Data Bank, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Longgang Shao
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhang
- Department of Prosthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jinjuan Wang
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Jianning Zhao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhongyang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Orthopedics, Air Force Hospital of Eastern Theater, Anhui Medical University, Nanjing, China.
| |
Collapse
|
4
|
Alrumaihi F, Almatroodi SA, Alharbi HOA, Alwanian WM, Alharbi FA, Almatroudi A, Rahmani AH. Pharmacological Potential of Kaempferol, a Flavonoid in the Management of Pathogenesis via Modulation of Inflammation and Other Biological Activities. Molecules 2024; 29:2007. [PMID: 38731498 PMCID: PMC11085411 DOI: 10.3390/molecules29092007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Natural products and their bioactive compounds have been used for centuries to prevent and treat numerous diseases. Kaempferol, a flavonoid found in vegetables, fruits, and spices, is recognized for its various beneficial properties, including its antioxidant and anti-inflammatory potential. This molecule has been identified as a potential means of managing different pathogenesis due to its capability to manage various biological activities. Moreover, this compound has a wide range of health-promoting benefits, such as cardioprotective, neuroprotective, hepatoprotective, and anti-diabetic, and has a role in maintaining eye, skin, and respiratory system health. Furthermore, it can also inhibit tumor growth and modulate various cell-signaling pathways. In vivo and in vitro studies have demonstrated that this compound has been shown to increase efficacy when combined with other natural products or drugs. In addition, kaempferol-based nano-formulations are more effective than kaempferol treatment alone. This review aims to provide detailed information about the sources of this compound, its bioavailability, and its role in various pathogenesis. Although there is promising evidence for its ability to manage diseases, it is crucial to conduct further investigations to know its toxicity, safety aspects, and mechanism of action in health management.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Hajed Obaid A. Alharbi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Wanian M. Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Fadiyah A. Alharbi
- Department of Obstetrics/Gynecology, Maternity and Children’s Hospital, Buraydah 52384, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
5
|
Zhang Z, Xu W, Zhang Z, Chen X, Jin H, Jiang N, Xu H. The bone-protective benefits of kaempferol combined with metformin by regulation of osteogenesis-angiogenesis coupling in OVX rats. Biomed Pharmacother 2024; 173:116364. [PMID: 38447449 DOI: 10.1016/j.biopha.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
This study was to investigate the potential mechanisms of treatment with metformin (Met) combined with kaempferol (Kae) against postmenopausal osteoporosis. Experiments were conducted in both ovariectomy (OVX)-induced osteoporosis rats and in vitro using RAW264.7 cells, MC3T3-E1 cells, and HUVECs. Results demonstrated the therapeutic effect of Met combined with Kae on osteoporosis. In vivo, Kae alone and in combination with Met treatments enhanced tibial trabecular microstructure, bone mineral density (BMD), and mechanical properties in OVX rats without causing hepatotoxicity and nephrotoxicity. It also reduced bone resorption markers (CTX-1 and TRAP) and increased the bone formation marker (PINP) level in the serum of OVX rats. The expression of bone resorption marker TRAP was reduced, while bone formation markers Runx2 and ALP were enhanced in the bone tissue of OVX rats. Furthermore, Met combined with Kae also promoted the expression of angiogenesis-related markers CD31 and VEGF in OVX rats. In vitro, MC3T3-E1s cells treated with Met combined with Kae showed higher expression of ALP, Runx2, and VEGF. Interestingly, the treatment did not directly promote HUVECs migration and angiogenesis, but enhanced osteoblast-mediated angiogenesis by upregulating VEGF levels. Additionally, Met combined with Kae treatment promoted VEGF secretion in MC3T3-E1, and activated the Notch intracelluar pathway by upregulating HES1 and HEY1 in HUVECs. Meantime, their stimulation on CD31 expression were inhibited by DAPT, a Notch signaling inhibitor. Overall, this study demonstrates the positive effects of Met combined with Kae on osteoporotic rats by promoting osteogenesis-angiogenesis coupling, suggesting their potential application in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Zhongyuan Zhang
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Wenshu Xu
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Zhenhua Zhang
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Xiaoxue Chen
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Hui Jin
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Ningning Jiang
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Hui Xu
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, People's Republic of China.
| |
Collapse
|
6
|
Zheng Y, Wang J, Xu K, Chen X. Intake of dietary flavonoids in relation to bone loss among U.S. adults: a promising strategy for improving bone health. Food Funct 2024; 15:766-778. [PMID: 38126227 DOI: 10.1039/d3fo02065g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Dietary flavonoids have been recommended for improving bone health due to their antioxidant, anti-inflammatory and osteogenic properties. However, the effectiveness of each flavonoid subclass in the prevention and treatment of osteoporosis remains controversial. The objective of the current study was to examine the association between the intake of flavonoid subclasses and bone loss in 10 480 U.S. adults in the National Health and Nutrition Examination Survey. We employed a multinomial logistic regression model to calculate the odds ratios (OR) and 95% confidence intervals (95% CI). The intake of flavones, isoflavones, and flavanones was beneficially associated with osteoporosis (ORQ5 vs. Q1 = 0.44; 95% CI: 0.30-0.64 for flavones; ORQ5 vs. Q1 = 0.53; 95% CI: 0.37-0.77 for isoflavones; ORQ5 vs. Q1 = 0.66; 95% CI: 0.45-0.97 for flavanones). A higher intake of flavones and flavanones was significantly associated with a lower risk of bone loss at the femoral neck rather than the lumbar spine. Notably, stratified analysis showed that genistein had a harmful association with osteopenia in the population with lower serum calcium levels, whereas it had a beneficial association with osteoporosis in the population with higher serum calcium levels. Multiple sensitivity analyses were performed to test the robustness of the results, including subgroup analysis, exclusion of individuals' use of anti-osteoporosis, corticosteroid, and estrogenic medications, adjusting more potential confounders and calculation of the E-value. Overall, incorporating this modifiable diet into an individual's lifestyle could provide potential possibilities to prevent and ameliorate osteoporosis.
Collapse
Affiliation(s)
- Yi Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jiacheng Wang
- Department of Epidemiology, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China
| |
Collapse
|
7
|
Yu X, Wu Q, Ren Z, Chen B, Wang D, Yuan T, Ding H, Wang Y, Yuan G, Wang Y, Zhang L, Zhao J, Sun Z. Kaempferol attenuates wear particle-induced inflammatory osteolysis via JNK and p38-MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117019. [PMID: 37574017 DOI: 10.1016/j.jep.2023.117019] [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: 02/03/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wear particle-induced inflammatory osteoclast activation is a master contributor to periprosthetic osteolysis, which can cause pathological bone loss and destruction. Hence, inhibiting inflammation and osteoclastogenesis is an important strategy for preventing wear particle-induced osteolysis. To date, there are no FDA-approved non-surgical pharmacotherapies for arresting periprosthetic osteolysis. Kaempferol (KAE), a natural flavonol abundant in many traditional Chinese herbal medicines, has been shown to have protective effects against inflammatory bone diseases such as rheumatoid arthritis, but no previous study has evaluated the effects of KAE on wear particle-induced osteolysis. AIM OF THE STUDY The study aimed to investigate the effects of KAE on wear particle-induced inflammatory osteolysis and osteoclast activation, and further explore the underlying mechanisms. MATERIALS AND METHODS TiAl6V4 metal particles (TiPs) were retrieved from the prosthesis of patients who underwent revision hip arthroplasty due to aseptic loosening. A mouse calvarial osteolysis model was used to investigate the effects of KAE on wear particle-induced inflammatory osteolysis in vivo. Primary bone marrow-derived macrophages (BMMs) were used to explore the effects of KAE on osteoclast differentiation and bone-resorbing activity as well as the underlying mechanisms in vitro. RESULTS In the present study, we found that KAE alleviated wear particle-induced inflammatory bone loss in vivo and inhibited osteoclast differentiation and function in vitro. Furthermore, we revealed that KAE exerted anti-osteoclastogenic effects by downregulating JNK and p38-MAPK signaling as well as the downstream NFATc1 expression. CONCLUSIONS KAE is an alternative therapeutic agent for preventing and treating periprosthetic osteolysis and aseptic loosening.
Collapse
Affiliation(s)
- Xin Yu
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Qi Wu
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China; Department of Vascular Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210023, China
| | - Bin Chen
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Dongsheng Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Tao Yuan
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Hao Ding
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yang Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Guodong Yuan
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuxiang Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Lei Zhang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jianning Zhao
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Zhongyang Sun
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China; Department of Orthopedics, Air Force Hospital of Eastern Theater, Anhui Medical University, Nanjing, 210002, China.
| |
Collapse
|
8
|
Cao N, Shou Z, Xiao Y, Liu P. Efficacy and Possible Mechanisms of Astragali Radix and its Ingredients in Animal Models of Osteoporosis: A Preclinical Review and Metaanalysis. Curr Drug Targets 2024; 25:135-148. [PMID: 38213165 DOI: 10.2174/0113894501275292231220062838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Astragali Radix (AR) has a long history as a traditional Chinese medicine for anti-osteoporosis (OP) treatment. The aim of the study was to explore the effect and optimal regimens of AR and its main ingredients (IAR) in OP treatment. METHODS Eligible animal studies were searched in seven databases (PubMed, Web of Science, MEDLINE, SciELO Citation Index, Cochrane Library, China National Knowledge Infrastructure and Wanfang). The primary outcomes were bone metabolic indices. The secondary outcome measure was the anti-OP mechanism of IAR. RESULTS 21 studies were enrolled in the study. The primary findings of the present article illustrated that IAR could significantly increase the bone mineral density (BMD), bone volume over the total volume, trabecular number, trabecular thickness, bone maximum load and serum calcium, while trabecular separation and serum C-terminal telopeptide of type 1 collagen were remarkably decreased (P < 0.05). In subgroup analysis, the BMD in the long treatment group (≥ 10 weeks) showed better effect size than the short treatment group (< 10 weeks) (P < 0.05). Modeling methods and animal sex were factors affecting serum alkaline phosphatase and osteocalcin levels. CONCLUSION The findings suggest the possibility of developing IAR as a drug for the treatment of OP. IAR with longer treatment time may achieve better effects regardless of animal strain and age.
Collapse
Affiliation(s)
- Ning Cao
- Pharmacy Department, The Second Affiliated Hospital, Zhejiang Chinese Medical University, China
| | - Zhangxuan Shou
- Pharmacy Department, The Second Affiliated Hospital, Zhejiang Chinese Medical University, China
| | - Yi Xiao
- HD Biosciences (A WuXi company) Pharma Tech, Shanghai 201201, China
| | - Puqing Liu
- Pharmacy Department, The Second Affiliated Hospital, Zhejiang Chinese Medical University, China
| |
Collapse
|
9
|
Zhang Y, Yang M, Li N, Li Q, Li Y, Zhai Y. Total Flavonoids Isolated from the Leaves of Eucommia ulmoides Augment Peak Bone Mass in Female Rats and Show no Side Effects in Other Organs. Curr Pharm Des 2024; 30:2410-2423. [PMID: 38963117 DOI: 10.2174/0113816128298755240613100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Eucommia ulmoides is a unique monophyletic and tertiary relict in China and is listed as a national second-class precious protected tree species. Eucommia ulmoides, recognized as a traditional Chinese medicine, can tonify the liver and kidneys and strengthen bones and muscles. Modern pharmacological research has proved that Eucommia ulmoides has multiple osteoprotective effects, including prohibiting the occurrence of osteoporosis and arthritis and enhancing the healing of bone fractures and bone defects. AIM To check its osteotropic effects, which may provide ideas for its potential use for the development of novel drugs to treat osteoporosis, this study evaluated the effect of total flavonoids from Eucommia ulmoides leaves (TFEL) on the acquisition of Peak Bone Mass (PBM) in young female rats. MATERIALS AND METHODS TFEL was isolated, and its purity was confirmed by using a UV spectrophotometer. TFEL with a purity of 85.09% was administered to 6-week-old female rats by oral gavage at a low (50), mid (100), or high (200 mg/kg/d) dose, and the control group was administrated only with the same volume of water. After 13 weeks of treatment, the rats were sacrificed, and serum, different organs, and limb bones (femurs and tibias) were harvested, and the bone turnover markers, organ index, Bone Mineral Density (BMD), biomechanical property, and microstructure parameters were assayed. Furthermore, molecular targets were screened, and network pharmacology analyses were conducted to reveal the potential mechanisms of action of TFEL. RESULTS Oral administration of TFEL for 13 weeks decreased the serum level of bone resorption marker TRACP-5b. As revealed by micro-computer tomography analysis, it elevated BMD even at a low dose (50 mg/kg/d) and improved the microstructural parameters, which were also confirmed by H&E histological staining. However, TFEL showed no effects on body weights, organ index, and micromorphology in the uterus. In our network pharmacology study, an intersection analysis screened out 64 shared targets, with quercetin, kaempferol, naringenin, and apigenin regulating the greatest number of targets associated with osteoporosis. Flavonoids in Eucommia ulmoides inhibited the occurrence of osteoporosis potentially through targeting signaling pathways for calcium, VEGF, IL-17, and NF-κB. Furthermore, AKT1, EGFR, PTGS2, VEGFA, and CALM were found to be potentially important target genes for the osteoprotective effects of flavonoids in Eucommia ulmoides. CONCLUSION The above results suggested that TFEL can be used to elevate the peak bone mass in adolescence in female individuals, which may prevent the occurrence of postmenopausal osteoporosis, and the good safety of TFEL also suggests that it can be used as a food additive for daily life to improve the bone health.
Collapse
Affiliation(s)
- Yun Zhang
- School of Stomatology, Henan University, Henan Kaifeng, 475000, China
- School of Pharmacy, Henan University, Henan Kaifeng, 475000, China
| | - Mingzhen Yang
- School of Stomatology, Henan University, Henan Kaifeng, 475000, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, School of Stomatology, Henan University, Henan Kaifeng, 475000, China
| | - Ningli Li
- School of Stomatology, Henan University, Henan Kaifeng, 475000, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, School of Stomatology, Henan University, Henan Kaifeng, 475000, China
| | - Qin Li
- School of Pharmacy, Henan University, Henan Kaifeng, 475000, China
| | - Yingying Li
- Department of Orthopaedics, Luoyang Orthopedic-Traumatological Hospital, Henan Luoyang, 471000, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Henan Kaifeng, 475000, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, School of Stomatology, Henan University, Henan Kaifeng, 475000, China
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Henan Kaifeng, 475000, China
| |
Collapse
|
10
|
Li S, Cui Y, Li M, Zhang W, Sun X, Xin Z, Li J. Acteoside Derived from Cistanche Improves Glucocorticoid-Induced Osteoporosis by Activating PI3K/AKT/mTOR Pathway. J INVEST SURG 2023; 36:2154578. [DOI: 10.1080/08941939.2022.2154578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Shumei Li
- The General Ward, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yajie Cui
- Department of Clinical Pharmacy, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Min Li
- Endocrine Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Wenting Zhang
- Department of Clinical Pharmacy, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Xiaoxue Sun
- Drug Clinical Trial Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zhaoxu Xin
- Department of Orthopaedics, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Jing Li
- Drug Clinical Trial Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
11
|
Ranjbar FE, Farzad-Mohajeri S, Samani S, Saremi J, Khademi R, Dehghan MM, Azami M. Kaempferol-loaded bioactive glass-based scaffold for bone tissue engineering: in vitro and in vivo evaluation. Sci Rep 2023; 13:12375. [PMID: 37524784 PMCID: PMC10390521 DOI: 10.1038/s41598-023-39505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023] Open
Abstract
Due to the increasing prevalence of bone disorders among people especially in average age, the future of treatments for osseous abnormalities has been illuminated by scaffold-based bone tissue engineering. In this study, in vitro and in vivo properties of 58S bioactive glass-based scaffolds for bone tissue engineering (bare (B.SC), Zein-coated (C.SC), and Zein-coated containing Kaempferol (KC.SC)) were evaluated. This is a follow-up study on our previously published paper, where we synthesized 58S bioactive glass-based scaffolds coated with Kaempferol-loaded Zein biopolymer, and characterized from mostly engineering points of view to find the optimum composition. For this aim, in vitro assessments were done to evaluate the osteogenic capacity and biological features of the scaffolds. In the in vivo section, all types of scaffolds with/without bone marrow-derived stem cells (BMSC) were implanted into rat calvaria bone defects, and potential of bone healing was assessed using imaging, staining, and histomorphometric analyses. It was shown that, Zein-coating covered surface cracks leading to better mechanical properties without negative effect on bioactivity and cell attachment. Also, BMSC differentiation proved that the presence of Kaempferol caused higher calcium deposition, increased alkaline phosphatase activity, bone-specific gene upregulation in vitro. Further, in vivo study confirmed positive effect of BMSC-loaded KC.SC on significant new bone formation resulting in complete bone regeneration. Combining physical properties of coated scaffolds with the osteogenic effect of Kaempferol and BMSCs could represent a new strategy for bone regeneration and provide a more effective approach to repairing critical-sized bone defects.
Collapse
Affiliation(s)
- Faezeh Esmaeili Ranjbar
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Saeed Farzad-Mohajeri
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Dr. Qarib Street, Azadi Street, Tehran, 1419963111, Iran
| | - Saeed Samani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, No. 88, Italia St., Keshavarz Blv, Tehran, Iran
| | - Jamileh Saremi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Rahele Khademi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, No. 88, Italia St., Keshavarz Blv, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Dr. Qarib Street, Azadi Street, Tehran, 1419963111, Iran.
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, No. 88, Italia St., Keshavarz Blv, Tehran, Iran.
| |
Collapse
|
12
|
Hanga-Farcaș A, Miere (Groza) F, Filip GA, Clichici S, Fritea L, Vicaș LG, Marian E, Pallag A, Jurca T, Filip SM, Muresan ME. Phytochemical Compounds Involved in the Bone Regeneration Process and Their Innovative Administration: A Systematic Review. PLANTS (BASEL, SWITZERLAND) 2023; 12:2055. [PMID: 37653972 PMCID: PMC10222459 DOI: 10.3390/plants12102055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 09/02/2023]
Abstract
Bone metabolism is a complex process which is influenced by the activity of bone cells (e.g., osteocytes, osteoblasts, osteoclasts); the effect of some specific biomarkers (e.g., parathyroid hormone, vitamin D, alkaline phosphatase, osteocalcin, osteopontin, osteoprotegerin, osterix, RANKL, Runx2); and the characteristic signaling pathways (e.g., RANKL/RANK, Wnt/β, Notch, BMP, SMAD). Some phytochemical compounds-such as flavonoids, tannins, polyphenols, anthocyanins, terpenoids, polysaccharides, alkaloids and others-presented a beneficial and stimulating effect in the bone regeneration process due to the pro-estrogenic activity, the antioxidant and the anti-inflammatory effect and modulation of bone signaling pathways. Lately, nanomedicine has emerged as an innovative concept for new treatments in bone-related pathologies envisaged through the incorporation of medicinal substances in nanometric systems for oral or local administration, as well as in nanostructured scaffolds with huge potential in bone tissue engineering.
Collapse
Affiliation(s)
- Alina Hanga-Farcaș
- Doctoral School of Biomedical Science, University of Oradea, 410087 Oradea, Romania;
| | - Florina Miere (Groza)
- Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (F.M.); (L.F.); (M.E.M.)
| | - Gabriela Adriana Filip
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babeș Street, 400347 Cluj-Napoca, Romania; (G.A.F.); (S.C.)
| | - Simona Clichici
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babeș Street, 400347 Cluj-Napoca, Romania; (G.A.F.); (S.C.)
| | - Luminita Fritea
- Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (F.M.); (L.F.); (M.E.M.)
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (E.M.); (A.P.); (T.J.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (E.M.); (A.P.); (T.J.)
| | - Annamaria Pallag
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (E.M.); (A.P.); (T.J.)
| | - Tunde Jurca
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (E.M.); (A.P.); (T.J.)
| | - Sanda Monica Filip
- Department of Physics, Faculty of Informatics and Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania;
| | - Mariana Eugenia Muresan
- Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania; (F.M.); (L.F.); (M.E.M.)
| |
Collapse
|
13
|
Liu W, Chen W, Hu M, Wang G, Hu Y, He Q, Xu Y, Tan J, Wang H, Huo L. Bioinformatics analysis combined with molecular dynamics simulation validation to elucidate the potential molecular mechanisms of Jianshen Decoction for treatment of osteoporotic fracture. Medicine (Baltimore) 2023; 102:e33610. [PMID: 37083798 PMCID: PMC10118375 DOI: 10.1097/md.0000000000033610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/03/2023] [Indexed: 04/22/2023] Open
Abstract
Osteoporotic fracture (OPF) is a prevalent skeletal disease in the middle-aged and elderly. In clinical practice, Jianshen Decoction (JSD) has been used to treat OPFs. However, the specific effective components and mechanisms of JSD on OPF have not been explored. Therefore, this study used bioinformatics analysis combined with molecular dynamics simulation validation to explore the molecular mechanism of JSD treatment of OPF. Public databases (TCMSP, Batman TCM) were used to find the effective active components and corresponding target proteins of JSD (screening conditions: OB ≥ 30%, drug-likeness ≥ 0.18, half-life ≥ 4). Differentially expressed genes (DEGs) related to OPF lesions were obtained based on the gene expression omnibus database (screening conditions: adjust P value < .01, | log2 FC | ≥ 1.0). The BisoGenet plug-in and the CytoNCA plug-in of Cytoscape were used to derive the potential core target proteins of JSD in the treatment of OPF. The JSD active ingredient target interaction network and the JSD-OPF target protein core network were constructed using the Cytoscape software. In addition, the R language Bioconductor package and clusterProfiler package were used to perform gene ontology (GO)/Kyoto Encylopedia Of Genes And Genome (KEGG) enrichment analysis on core genes to explain the biological functions and signal pathways of core proteins. Finally, molecular docking and molecular dynamics simulations were carried out through PyMOL, AutoDockTools 1.5.6, Vina, LeDock, Discovery Studio (DS) 2019, and other software to verify the binding ability of drug active ingredients and core target proteins. A total of 245 targets and 70 active components were identified. Through protein-protein interaction (PPI) network construction, 39 core targets were selected for further research. GO/KEGG enrichment analysis showed that the DNA-binding transcription factor binding, RNA polymerase II-specific DNA-binding transcription factor binding, MAPK signaling pathway, and ErbB signaling pathway were mainly involved. The results of molecular docking and molecular dynamics simulations supported the good interaction between MYC protein and Quercetin/Stigmasterol. In this study, bioinformatics, molecular docking, and molecular dynamics simulations were used for the first time to clarify the active components, molecular targets, and key biological pathways of JSD in the treatment of OPF, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Weinian Liu
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weijian Chen
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengting Hu
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guangwei Wang
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuanhao Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qi He
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yidong Xu
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jun Tan
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial People’s Hospital’s Nanhai Hospital, Foshan, Guangdong, China
| | - Haibin Wang
- Department of Orthopaedics of the First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liwei Huo
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Moorthy T, Hathim B M, NagaMahesh C H M, Anburaj G, Ahmed SSSJ, Gopinath V, Munuswamy-Ramanujam G, Rao SK, Kamath MS. Controlled release of kaempferol from porous scaffolds augments in-vitro osteogenesis in human osteoblasts. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
15
|
Bioactivity, Molecular Mechanism, and Targeted Delivery of Flavonoids for Bone Loss. Nutrients 2023; 15:nu15040919. [PMID: 36839278 PMCID: PMC9960663 DOI: 10.3390/nu15040919] [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] [Received: 01/11/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Skeletal disabilities are a prominent burden on the present population with an increasing life span. Advances in osteopathy have provided various medical support for bone-related diseases, including pharmacological and prosthesis interventions. However, therapeutics and post-surgery complications are often reported due to side effects associated with modern-day therapies. Thus, therapies utilizing natural means with fewer toxic or other side effects are the key to acceptable interventions. Flavonoids constitute a class of bioactive compounds found in dietary supplements, and their pharmacological attributes have been well appreciated. Recently, flavonoids' role is gaining renowned interest for its effect on bone remodeling. A wide range of flavonoids has been found to play a pivotal role in the major bone signaling pathways, such as wingless-related integration site (Wnt)/β-catenin, bone morphogenetic protein (BMP)/transforming growth factor (TGF)-β, mitogen-activated protein kinase (MAPK), etc. However, the reduced bioavailability and the absorption of flavonoids are the major limitations inhibiting their use against bone-related complications. Recent utilization of nanotechnological approaches and other delivery methods (biomaterial scaffolds, micelles) to target and control release can enhance the absorption and bioavailability of flavonoids. Thus, we have tried to recapitulate the understanding of the role of flavonoids in regulating signaling mechanisms affecting bone remodeling and various delivery methods utilized to enhance their therapeutical potential in treating bone loss.
Collapse
|
16
|
Xu J, Ma J, Zeng Y, Si H, Wu Y, Zhang S, Shen B. Transcriptome-wide association study identifies novel genes associated with bone mineral density and lean body mass in children. Endocrine 2023; 79:400-409. [PMID: 36572794 PMCID: PMC9892108 DOI: 10.1007/s12020-022-03225-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/05/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To identify novel candidate genes whose expression is associated with bone mineral density (BMD) and body lean mass (LM) in children. METHODS A tissue-specific transcriptome-wide association study (TWAS) was conducted utilizing a large-scale genome-wide association study (GWAS) dataset associated with BMD and LM and involving 10,414 participants. The measurement of BMD and LM phenotypes was made based on total-body dual-energy X-ray absorptiometry (TB-DXA) scans. TWAS was conducted by using FUSION software. Reference panels for muscle skeleton (MS), peripheral blood (NBL) and whole blood (YBL) were used for TWAS analysis. Functional enrichment and protein-protein interaction (PPI) analyses of the genes identified by TWAS were performed by using the online tool Metascape ( http://metascape.org ). RESULTS For BMD, we identified 174 genes with P < 0.05, such as IKZF1 (P = 1.46 × 10-9) and CHKB (P = 8.31 × 10-7). For LM, we identified 208 genes with P < 0.05, such as COPS5 (P = 3.03 × 10-12) and MRPS33 (P = 5.45 × 10-10). Gene ontology (GO) enrichment analysis of the BMD-associated genes revealed 200 GO terms, such as protein catabolic process (Log P = -5.09) and steroid hormone-mediated signaling pathway (Log P = -3.13). GO enrichment analysis of the LM-associated genes detected 287 GO terms, such as the apoptotic signaling pathway (Log P = -8.08) and lipid storage (Log P = -3.55). CONCLUSION This study identified several candidate genes for BMD and LM in children, providing novel clues to the genetic mechanisms underlying the development of childhood BMD and LM.
Collapse
Affiliation(s)
- Jiawen Xu
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Jun Ma
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Yi Zeng
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Haibo Si
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Yuangang Wu
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Shaoyun Zhang
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Bin Shen
- Orthopedic Research Institute, Department of Orthopedics, Sichuan University West China Hospital, 37# Guoxue Road, Chengdu, 610041, Sichuan Province, People's Republic of China.
| |
Collapse
|
17
|
Network Pharmacological Analysis and Animal Experimental Study on Osteoporosis Treatment with GuBen-ZengGu Granules. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:9317557. [PMID: 36686973 PMCID: PMC9851784 DOI: 10.1155/2023/9317557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023]
Abstract
Aim We explored the molecular pathway and material basis of GuBen-ZengGu granules (GBZGG) in treating osteoporosis using network pharmacology and animal experiments. Methods The effective active components and potential targets of GBZGG were obtained from the TCMSP database and BATMAN-TCM database. Disease-related genes were obtained from GeneCard, NCBI, and DisGeNET. Next, a protein interaction network was established using the STRING database, and core genes were screened using the MCODE module. Cytoscape 3.8.0 was used to construct the network of component-disease-pathway-target, and KEGG pathway enrichment analyses were performed using the clusterProfiler R package to predict the mechanism of GBZGG in treating osteoporosis. An osteoporosis rat model was established by ovarian excision (OVX), and the partial results of network pharmacology were experimentally verified. Results Pharmacodynamic results showed that GBZGG increased bone mineral density (BMD) and significantly improved the indexes of femur microstructure in model rats. The network pharmacology results showed that quercetin, luteolin, stigmasterol, angelicin, kaempferol, bakuchiol, bakuchiol, 7-O-methylisomucronulatum, isorhamnetin, formononetin, and beta-sitosterol are the major components of GBZGG, with MAPK1, AKT1, JUN, HSP90AA1, RELA, MAPK14, ESR1, RXRA, FOS, MAPK8, NCOA1, MYC, and IL-6 as its core targets for treating osteoporosis. Biological effects could be exerted by regulating the signaling pathways of fluid shear stress and the signaling pathways of atherosclerosis, advanced glycation end products (AGE-RAGE) of diabetic complications, prostate cancer, interleukin (IL-17), tumor necrosis factor (TNF), hepatitis B, mitogen-activated protein kinase (MAPK), etc. The results of animal experiments showed that GBZGG could reduce the serum levels of IL-6 and TNF-α, increase the expression of bone morphogenetic protein-2 (BMP-2) and runt-related transcription factor 2 (RUNX2) protein, and inhibit the activity of extracellular-regulated protein kinases (ERK1/2) and phosphorylation ERK1/2 (p-ERK1/2) protein. Conclusion GBZGG reduces the expression of ERK1/2 and p-ERK1/2 proteins and mRNAs through the inhibitory effects on IL-6 and TNF-α and negatively regulates the MAPK/ERK signaling pathway. The osteoporosis model showed that it effectively improved the loss of bone mass and destruction of bone microstructure in rats and maintained a positive balance for bone metabolism.
Collapse
|
18
|
徐 鑫, 范 骁, 吴 鑫, 时 利, 王 培, 高 福, 孙 伟, 李 子. [Protective effect of Kaempferol on endothelial cell injury in glucocorticoid induced osteonecrosis of the femoral head]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:1277-1287. [PMID: 36310467 PMCID: PMC9626266 DOI: 10.7507/1002-1892.202204028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/23/2022] [Indexed: 01/24/2023]
Abstract
Objective To explore the effect of Kaempferol on bone microvascular endothelial cells (BMECs) in glucocorticoid induced osteonecrosis of the femoral head (GIONFH) in vitro. Methods BMECs were isolated from cancellous bone of femoral head or femoral neck donated voluntarily by patients with femoral neck fracture. BMECs were identified by von Willebrand factor and CD31 immunofluorescence staining and tube formation assay. The cell counting kit 8 (CCK-8) assay was used to screen the optimal concentration and the time point of dexamethasone (Dex) to inhibit the cell activity and the optimal concentration of Kaempferol to improve the inhibition of Dex. Then the BMECs were divided into 4 groups, namely, the cell group (group A), the cells treated with optimal concentration of Dex group (group B), the cells treated with optimal concentration of Dex+1 μmol/L Kaempferol group (group C), and the cells treated with optimal concentration of Dex+5 μmol/L Kaempferol group (group D). EdU assay, in vitro tube formation assay, TUNEL staining assay, Annexin Ⅴ/propidium iodide (PI) staining assay, Transwell migration assay, scratch healing assay, and Western blot assay were used to detect the effect of Kaempferol on the proliferation, tube formation, apoptosis, migration, and protein expression of BMECs treated with Dex. Results The cultured cells were identified as BMECs. CCK-8 assay showed that the optimal concentration and the time point of Dex to inhibit cell activity was 300 μmol/L for 24 hours, and the optimal concentration of Kaempferol to improve the inhibitory activity of Dex was 1 μmol/L. EdU and tube formation assays showed that the cell proliferation rate, tube length, and number of branch points were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). TUNEL and Annexin V/PI staining assays showed that the rates of TUNEL positive cells and apoptotic cells were significantly higher in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Scratch healing assay and Transwell migration assay showed that the scratch healing rate and the number of migration cells were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Western blot assay demonstrated that the relative expressions of Cleaved Caspase-3 and Bax proteins were significantly higher in groups B-D than in group A, and in groups B and D than in group C ( P<0.05); the relative expressions of matrix metalloproteinase 2, Cyclin D1, Cyclin E1, VEGFA, and Bcl2 proteins were significantly lower in groups B-D than in group A, and in groups B and D than in group C ( P<0.05). Conclusion Kaempferol can alleviate the damage and dysfunction of BMECs in GIONFH.
Collapse
Affiliation(s)
- 鑫 徐
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 骁宇 范
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 鑫杰 吴
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 利军 时
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 培旭 王
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 福强 高
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| | - 伟 孙
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
- 北京大学中日友好临床医学院骨科(北京 100029)Department of Orthopedics, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, P. R. China
| | - 子荣 李
- 中日友好医院骨科 北京协和医学院研究生院 中国医学科学院(北京 100029)Department of Orthopedics, China-Japan Friendship Hospital, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, P. R. China
| |
Collapse
|
19
|
Research on the Mechanism of Liuwei Dihuang Decoction for Osteoporosis Based on Systematic Biological Strategies. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7017610. [PMID: 36185080 PMCID: PMC9522519 DOI: 10.1155/2022/7017610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 02/21/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022]
Abstract
Background Osteoporosis is an important health problem worldwide. Liuwei Dihuang Decoction (LDD) and its main ingredients may have a good clinical effect on osteoporosis. Meanwhile, its mechanism for treating osteoporosis needs to be further revealed in order to provide a basis for future drug development. Methods A systematic biological methodology was utilized to construct and analyze the LDD-osteoporosis network. After that, the human transcription data of LDD intervention in patients with osteoporosis and protein arrays data of LDD intervention in osteoporosis rats were collected. The human transcription data analysis, protein arrays data analysis, and molecular docking were performed to validate the findings of the prediction network (LDD-osteoporosis PPI network). Finally, animal experiments were conducted to verify the prediction results of systematic pharmacology. Results (1) LDD-osteoporosis PPI network shows the potential compounds, potential targets (such as ALB, IGF1, SRC, and ESR1), clusters, biological processes (such as positive regulation of calmodulin 1-monooxygenase activity, estrogen metabolism, and endothelial cell proliferation), and signaling and Reactome pathways (such as JAK-STAT signaling pathway, osteoclast differentiation, and degradation of the extracellular matrix) of LDD intervention in osteoporosis. (2) Human transcriptomics data and protein arrays data validated the findings of the LDD-osteoporosis PPI network. (3) The animal experiments showed that LDD can improve bone mineral density (BMD), increase serum estradiol (E2) and alkaline phosphatase (ALP) levels, and upregulate Wnt3a and β-catenin mRNA expression (P < 0.05). (4) Molecular docking results showed that alisol A, dioscin, loganin, oleanolic acid, pachymic acid, and ursolic acid may stably bind to JAK2, ESR1, and CTNNB1. Conclusion LDD may have a therapeutic effect on osteoporosis through regulating the targets (such as ALB, IGF1, SRC, and ESR1), biological processes (such as positive regulation of calmodulin 1-monooxygenase activity, estrogen metabolism, and endothelial cell proliferation), and pathways (such as JAK-STAT signaling pathway, osteoclast differentiation, and degradation of the extracellular matrix) found in this research.
Collapse
|
20
|
Rai R, Singh KB, Khanka S, Maurya R, Singh D. Cladrin alleviates dexamethasone-induced apoptosis of osteoblasts and promotes bone formation through autophagy induction via AMPK/mTOR signaling. Free Radic Biol Med 2022; 190:339-350. [PMID: 35998794 DOI: 10.1016/j.freeradbiomed.2022.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/05/2022] [Accepted: 08/16/2022] [Indexed: 12/09/2022]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is a common clinical consequence that arises due to the extensive usage of glucocorticoids. Cladrin (Clad), a methoxylated isoflavone has been reported to have a bone protecting effect by enhancing osteoblast proliferation and differentiation. However, its consequences on GIOP are not reported yet. This study investigates whether Clad protects against the deleterious effects of Dexamethasone (Dex) on osteoblast and bone. Mice calvarial osteoblasts were treated with Clad and then exposed to Dex to study the effect on osteoblast differentiation, proliferation, and survival. Further, GIOP mice were treated with Clad (5 and 10 mg/kg) doses along with reference standard alendronate (ALN 3 mg/kg) for evaluation of bone protecting effect of Clad. We analyzed bone and vertebral microarchitecture, mechanical strength, and biochemical parameters. We observed that Clad at 10 nM concentration mitigated Dex-induced cytotoxicity and defend osteoblasts against apoptosis. Subsequent results demonstrate that Clad suppressed apoptosis of osteoblast in the presence of Dex by enhancing autophagy in a way that was reliant on the AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) pathway. Furthermore, micro-CT scanning, eco MRI results, and serum CTX levels revealed that 12 weeks of Clad treatment prevented bone loss and preserved trabecular bone mass in GIOP animals. We also observed that Clad treated osteoblasts had a lower rate of apoptosis and a greater LC3-II/LC3-I ratio than the Dex group. Our findings show that Clad can protect osteoblasts against glucocorticoids by inducing autophagy via the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Reena Rai
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Krishna Bhan Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Uttar Pradesh, 201002, India
| | - Sonu Khanka
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Uttar Pradesh, 201002, India
| | - Rakesh Maurya
- Division of Medicinal & Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Divya Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
21
|
Soluble Free, Esterified and Insoluble-Bound Phenolic Antioxidants from Chickpeas Prevent Cytotoxicity in Human Hepatoma HuH-7 Cells Induced by Peroxyl Radicals. Antioxidants (Basel) 2022; 11:antiox11061139. [PMID: 35740036 PMCID: PMC9219979 DOI: 10.3390/antiox11061139] [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: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Chickpeas are rich sources of bioactive compounds such as phenolic acids, flavonoids, and isoflavonoids. However, the contribution of insoluble-bound phenolics to their antioxidant properties remains unclear. Four varieties of chickpeas were evaluated for the presence of soluble (free and esterified) and insoluble-bound phenolics as well as their antiradical activity, reducing power and inhibition of peroxyl-induced cytotoxicity in human HuH-7 cells. In general, the insoluble-bound fraction showed a higher total phenolic content. Phenolic acids, flavonoids, and isoflavonoids were identified and quantified by UPLC-MS/MS. Taxifolin was identified for the first time in chickpeas. However, m-hydroxybenzoic acid, taxifolin, and biochanin A were the main phenolics found. Biochanin A was mostly found in the free fraction, while m-hydroxybenzoic acid was present mainly in the insoluble-bound form. The insoluble-bound fraction made a significant contribution to the reducing power and antiradical activity towards peroxyl radical. Furthermore, all extracts decreased the oxidative damage of human HuH-7 cells induced by peroxyl radicals, thus indicating their hepatoprotective potential. This study demonstrates that the antioxidant properties and bioactive potential of insoluble-bound phenolics of chickpeas should not be neglected.
Collapse
|
22
|
Wang A, Yuan W, Song Y, Zang Y, Yu Y. Osseointegration Effect of Micro-Nano Implants Loaded With Kaempferol in Osteoporotic Rats. Front Bioeng Biotechnol 2022; 10:842014. [PMID: 35284417 PMCID: PMC8905647 DOI: 10.3389/fbioe.2022.842014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/21/2022] [Indexed: 01/22/2023] Open
Abstract
Objective: To investigate the effect of osseointegration of kaempferol loaded on the surface of micro-nanomorphic implants in ovariectomized rats. Methods: Titanium flakes were polished to obtain the PT group, anodized and acid-etched to obtain the NT and WNT groups, loaded with kaempferol to obtain the KNT and KWNT groups, and spin-coated on chitosan-gelatin composite film to obtain the KNT-CG and KWNT-CG groups. In vitro experiments were performed to observe the physicochemical properties of the titanium tablets in each group through scanning electron microscopy and contact angle experiments. The cytotoxicity and drug release pattern were observed using CCK-8 and drug release assays. An osteoporosis rat model was established. Pure titanium implants were divided into PT, NT, WNT, KNT-CG, and KWNT-CG groups after the same treatment and used in the in vivo experiments and then implanted in the femur of mice in each group. After 4 weeks, all samples were collected for toluidine blue staining, micro-computed tomography scanning, and bone morphometry analysis to evaluate their osteogenic properties. Results: According to scanning electron microscopy, the surface of the titanium flakes had a micro-nano morphology in the WNT group and the KNT and KWNT groups were functionally loaded with kaempferol. In CCK-8 and drug release experiments, the loaded kaempferol and gelatin composite membranes showed no significant toxic effects on cells. The drug release time in the KNT-CG and KWNT-CG groups was significantly longer than that in the KNT and KWNT groups, with the release time in the KWNT-CG group reaching 15 days. In vivo experiments micro-computed tomography and bone morphometry analysis showed that the osteoporosis model had been successfully constructed. The bone volume fraction around the implant increased. Toluidine blue staining showed new bone formation and a significantly increased number of bone trabeculae. Conclusion: Kaempferol micro-nanocomposite coating improved the osseointegration ability of implants in osteoporotic rats.
Collapse
Affiliation(s)
- Anyue Wang
- Department of Stomatology, School of Stomatology of Qingdao University, Qingdao, China
| | | | - Yu Song
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yanjun Zang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yanling Yu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
- *Correspondence: Yanling Yu,
| |
Collapse
|
23
|
Sekaran S, Thangavelu L. Re-appraising the role of flavonols, flavones and flavonones on osteoblasts and osteoclasts- A review on its molecular mode of action. Chem Biol Interact 2022; 355:109831. [PMID: 35120918 DOI: 10.1016/j.cbi.2022.109831] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/02/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
Bone disorders have become a global concern illustrated with decreased bone mineral density and disruption in microarchitecture of natural bone tissue organization. Natural compounds that promote bone health by augmenting osteoblast functions and suppressing osteoclast functions has gained much attention and offer greater therapeutic value compared to conventional therapies. Amongst several plant-based molecules, flavonoids act as a major combatant in promoting bone health through their multi-faceted biological activities such as antioxidant, anti-inflammatory, and osteogenic properties. They protect bone loss by regulating the signalling cascades involved in osteoblast and osteoclast functions. Flavonoids augment osteoblastogenesis and inhibits osteoclastogenesis through their modulation of various signalling pathways. This review discusses the role of various flavonoids and their molecular mechanisms involved in maintaining bone health by regulating osteoblast and osteoclast functions.
Collapse
Affiliation(s)
- Saravanan Sekaran
- Centre for Trans-disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute for Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India.
| | - Lakshmi Thangavelu
- Centre for Trans-disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute for Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| |
Collapse
|
24
|
Hemp Seeds in Post-Arthroplasty Rehabilitation: A Pilot Clinical Study and an In Vitro Investigation. Nutrients 2021; 13:nu13124330. [PMID: 34959882 PMCID: PMC8709006 DOI: 10.3390/nu13124330] [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: 10/07/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis is a type of degenerative joint disease that results from the breakdown of joint cartilage and underlying bone. Due to their antioxidants and anti-inflammatory action, the phytochemical constituents of many vegetable varieties could represent a new frontier for the treatment of patients with Osteoarthritis and are still being explored. The aim of this pilot human study was to investigate the effects of pasta enriched with hemp seed flour on osteoarticular pain and bone formation markers in patients in post-arthroplasty rehabilitation. Another purpose was to evaluate the effect of hemp seed extract on bone metabolism, in vitro. A pilot, controlled, clinical study was conducted to verify the feasibility of pain symptom reduction in patients with Osteoarthritis undergoing arthroplasty surgery. We also investigated the effect of hemp seed extract on the Wnt/β-catenin and ERK1/2 pathways, alkaline phosphatase, RANKL, RUNX-2, osteocalcin, and COL1A on Saos-2. After 6 weeks, the consumption of hemp seed pasta led to greater pain relief compared to the regular pasta control group (−2.9 ± 1.3 cm vs. −1.3 ± 1.3 cm; p = 0.02). A significant reduction in serum BALP was observed in the participants consuming the hemp seed pasta compared to control group (−2.8 ± 3.2 µg/L vs. 1.1 ± 4.3 µg/L; p = 0.04). In the Saos-2 cell line, hemp seed extract also upregulated Wnt/β-catenin and Erk1/2 pathways (p = 0.02 and p = 0.03) and osteoblast differentiation markers (e.g., ALP, OC, RUNX2, and COL1A) and downregulated RANKL (p = 0.02), compared to the control. Our study demonstrated that hemp seed can improve pain symptoms in patients with osteoarthritis undergoing arthroplasty surgery and also improves bone metabolism both in humans and in vitro. However, more clinical studies are needed to confirm our preliminary findings.
Collapse
|
25
|
Xie B, Zeng Z, Liao S, Zhou C, Wu L, Xu D. Kaempferol Ameliorates the Inhibitory Activity of Dexamethasone in the Osteogenesis of MC3T3-E1 Cells by JNK and p38-MAPK Pathways. Front Pharmacol 2021; 12:739326. [PMID: 34675808 PMCID: PMC8524096 DOI: 10.3389/fphar.2021.739326] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022] Open
Abstract
Kaempferol has been reported to exhibit beneficial effect on the osteogenic differentiation in mesenchymal stem cells (MSC) and osteoblasts. In our previous study, dexamethasone (DEX) demonstrated inhibitory effect on MC3T3-E1 cells differentiation. In this study, we mainly explored the protective effect of kaempferol on the inhibitory activity of DEX in the osteogenesis of MC3T3-E1 cells. We found that kaempferol ameliorated the proliferation inhibition, cell cycle arrest, and cell apoptosis and increased the activity of alkaline phosphatase (ALP) and the mineralization in DEX-treated MC3T3-E1 cells. Kaempferol also significantly enhanced the expression of osterix (Osx) and runt-related transcription factor 2 (Runx2) in MC3T3-E1 cells treated with DEX. In addition, kaempferol attenuated DEX-induced reduction of cyclin D1 and Bcl-2 expression and elevation of p53 and Bax expression. Kaempferol also activated JNK and p38-MAPK pathways in DEX-treated MC3T3-E1 cells. Furthermore, kaempferol improved bone mineralization in DEX-induced bone damage in a zebrafish larvae model. These data suggested that kaempferol ameliorated the inhibitory activity of DEX in the osteogenesis of MC3T3-E1 cells by activating JNK and p38-MAPK signaling pathways. Kaempferol exhibited great potentials in developing new drugs for treating glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Baocheng Xie
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Zhanwei Zeng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Shiyi Liao
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Chenhui Zhou
- School of Nursing, Guangdong Medical University, Dongguan, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Daohua Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| |
Collapse
|
26
|
Seely KD, Kotelko CA, Douglas H, Bealer B, Brooks AE. The Human Gut Microbiota: A Key Mediator of Osteoporosis and Osteogenesis. Int J Mol Sci 2021; 22:9452. [PMID: 34502371 PMCID: PMC8431678 DOI: 10.3390/ijms22179452] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/14/2022] Open
Abstract
An expanding body of research asserts that the gut microbiota has a role in bone metabolism and the pathogenesis of osteoporosis. This review considers the human gut microbiota composition and its role in osteoclastogenesis and the bone healing process, specifically in the case of osteoporosis. Although the natural physiologic processes of bone healing and the pathogenesis of osteoporosis and bone disease are now relatively well known, recent literature suggests that a healthy microbiome is tied to bone homeostasis. Nevertheless, the mechanism underlying this connection is still somewhat enigmatic. Based on the literature, a relationship between the microbiome, osteoblasts, osteoclasts, and receptor activator of nuclear factor-kappa-Β ligand (RANKL) is contemplated and explored in this review. Studies have proposed various mechanisms of gut microbiome interaction with osteoclastogenesis and bone health, including micro-RNA, insulin-like growth factor 1, and immune system mediation. However, alterations to the gut microbiome secondary to pharmaceutical and surgical interventions cannot be discounted and are discussed in the context of clinical therapeutic consideration. The literature on probiotics and their mechanisms of action is examined in the context of bone healing. The known and hypothesized interactions of common osteoporosis drugs and the human gut microbiome are examined. Since dysbiosis in the gut microbiota can function as a biomarker of bone metabolic activity, it may also be a pharmacological and nutraceutical (i.e., pre- and probiotics) therapeutic target to promote bone homeostasis.
Collapse
Affiliation(s)
- Kevin D. Seely
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Cody A. Kotelko
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Hannah Douglas
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Brandon Bealer
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Amanda E. Brooks
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
- Department of Research and Scholarly Activity, Rocky Vista University, Ivins, UT 84738, USA
| |
Collapse
|
27
|
Cheng T, Cao J, Jiang X, Yarmolenko M, Rogachev A, Rogachev A. Study of Icaritin Films by Low-Energy Electron Beam Deposition. EURASIAN CHEMICO-TECHNOLOGICAL JOURNAL 2021. [DOI: 10.18321/ectj1077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this paper, icaritin film was prepared by low-energy beam electron beam deposition (EBD). The material test showed that the structure and composition of icaritin were not changed after electron beam deposition. Then, the film was sliced and immersed in simulated body fluids, it can be seen that the film was released quickly in the first 7 days. With the extension of soaking time, the release rate gradually slowed down, and the release amount exceeded 90% in about 20 days. In vitro cytotoxicity test showed that the relative cell viability rate of the film was still 92.32±1.30% (p<0.05), indicating that the film possessed excellent cytocompatibility.
Collapse
|
28
|
Imtiyaz Z, Lin YT, Liang FY, Chiou WF, Lee MH. Compounds Isolated from Wikstroemia taiwanensis Regulate Bone Remodeling by Modulating Osteoblast and Osteoclast Activities. Front Pharmacol 2021; 12:670254. [PMID: 34349644 PMCID: PMC8327267 DOI: 10.3389/fphar.2021.670254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
Bone remodeling, a dynamic process in which bone formation by osteoblast is preceded by bone resorption by osteoclast, is a vital physiological process for maintaining bone mass and strength, imbalances in which could precipitate osteoporosis. Due to the unilateral mechanism of the existing bone remodeling drugs, identifying compounds that could regulate the balance between osteoclast and osteoblast could improve the treatment of osteoporosis. Here, we show that compounds isolated from Wikstroemia taiwanensis modulate osteoclast and osteoblast activities. Specifically, astragalin (1) and kaempferol 3-O-β-D-apiofuranosyl-(1→6)-β-D-glucopyranoside (2), besides increasing mineral deposition, increased alkaline phosphatase activity (137.2% for 1 and 115.8% for 2) and ESR-α expression (112.8% for 1 and 122.5% for 2) in primary human osteoblasts. In contrast, compounds 1, 2, 3, and 5 inhibited tartrate-resistant acid phosphatase (TRAP) activity in receptor activator of nuclear factor-κB ligand-induced osteoclasts by 40.8, 17.1, 25.9, and 14.5% and also decreased the number of TRAP-positive cells by 51.6, 26.8, 20.5, and 18.6%, respectively. Our findings, therefore, showed that compounds isolated from W. taiwanensis could increase osteoblast activity while simultaneously decreasing osteoclast activity, and hence, warrant further evaluation for development as anti-osteoporosis agents.
Collapse
Affiliation(s)
- Zuha Imtiyaz
- PhD in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Yi-Tzu Lin
- PhD in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Fang-Yu Liang
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wen-Fei Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Mei-Hsien Lee
- PhD in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
29
|
Prakash R, Mishra T, Dev K, Sharma K, Kuldeep J, John AA, Tripathi A, Sharma C, Arya KR, Kumar B, Siddiqi MI, Tadigoppula N, Singh D. Phenanthrenoid Coelogin Isolated from Coelogyne cristata Exerts Osteoprotective Effect Through MAPK-Mitogen-Activated Protein Kinase Signaling Pathway. Calcif Tissue Int 2021; 109:32-43. [PMID: 33675370 DOI: 10.1007/s00223-021-00818-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
Osteoporosis is a major health problem in postmenopausal women globally. This study determined the mechanism through which coelogin stimulates osteoblastogenesis and its osteoprotective and bone regenerating potential. Coelogin effect on primary calvarial osteoblast cells was determined by measuring alkaline phosphatase activity, mineralization, osteoblast survival, and apoptosis and protein expression studies. The osteoprotective effect of coelogin was also evaluated on osteopenic adult female Swiss mice. At autopsy, bones were collected for dynamic and histomorphometry studies. Serum samples were also collected for assessment of serum parameters. Coelogin treatment led to increased osteoblast proliferation, survival, differentiation, and mineralization in osteoblast cells. Coelogin supplementation to Ovx mice promoted new bone formation, prevented Ovx-induced deterioration of bone microarchitecture, and enhanced bone regeneration. In addition, signaling studies revealed that coelogin treatment activates the ER-Erk and Akt-dependent signaling pathways which stimulate the osteoblastogenesis in osteoblast cells.
Collapse
Affiliation(s)
- Ravi Prakash
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Tripti Mishra
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Kapil Dev
- Ethnobotany Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Kriti Sharma
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Jitendra Kuldeep
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Aijaz Ahmad John
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Alok Tripathi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Chetan Sharma
- Ethnobotany Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Kamal Ram Arya
- Ethnobotany Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Brijesh Kumar
- Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Mohd Imran Siddiqi
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Narender Tadigoppula
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India.
| | - Divya Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
30
|
Zhao B, Peng Q, Poon EHL, Chen F, Zhou R, Shang G, Wang D, Xu Y, Wang R, Qi S. Leonurine Promotes the Osteoblast Differentiation of Rat BMSCs by Activation of Autophagy via the PI3K/Akt/mTOR Pathway. Front Bioeng Biotechnol 2021; 9:615191. [PMID: 33708763 PMCID: PMC7940513 DOI: 10.3389/fbioe.2021.615191] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023] Open
Abstract
Background Leonurine, a major bioactive component from Herba leonuri, has been shown to exhibit anti-inflammatory and antioxidant effects. The aim of this study was to investigate the effect of leonurine on bone marrow-derived mesenchymal stem cells (BMSCs) as a therapeutic approach for treating osteoporosis. Materials and Methods Rat bone marrow-derived mesenchymal stem cells (rBMSCs) were isolated from 4-weeks-old Sprague–Dawley rats. The cytocompatibility of leonurine on rBMSCs was tested via CCK-8 assays and flow cytometric analyses. The effects of leonurine on rBMSC osteogenic differentiation were analyzed via ALP staining, Alizarin red staining, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot. Additionally, autophagy-related markers were examined via qRT-PCR and Western blot analyses of rBMSCs during osteogenic differentiation with leonurine and with or without 3-methyladenine (3-MA) as an autophagic inhibitor. Finally, the PI3K/Akt/mTOR signaling pathway was evaluated during rBMSC osteogenesis. Results Leonurine at 2–100 μM promoted the proliferation of rBMSCs. ALP and Alizarin red staining results showed that 10 μM leonurine promoted rBMSC osteoblastic differentiation, which was consistent with the qRT-PCR and Western blot results. Compared with those of the control group, the mRNA and protein levels of Atg5, Atg7, and LC3 were upregulated in the rBMSCs upon leonurine treatment. Furthermore, leonurine rescued rBMSC autophagy after inhibition by 3-MA. Additionally, the PI3K/AKT/mTOR pathway was activated in rBMSCs upon leonurine treatment. Conclusion Leonurine promotes the osteoblast differentiation of rBMSCs by activating autophagy, which depends on the PI3K/Akt/mTOR pathway. Our results suggest that leonurine may be a potential treatment for osteoporosis.
Collapse
Affiliation(s)
- Bingkun Zhao
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qian Peng
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Enoch Hin Lok Poon
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Fubo Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rong Zhou
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guangwei Shang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Raorao Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengcai Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
31
|
Pérez-Lozano ML, Cesaro A, Mazor M, Esteve E, Berteina-Raboin S, Best TM, Lespessailles E, Toumi H. Emerging Natural-Product-Based Treatments for the Management of Osteoarthritis. Antioxidants (Basel) 2021; 10:265. [PMID: 33572126 PMCID: PMC7914872 DOI: 10.3390/antiox10020265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is a complex degenerative disease in which joint homeostasis is disrupted, leading to synovial inflammation, cartilage degradation, subchondral bone remodeling, and resulting in pain and joint disability. Yet, the development of new treatment strategies to restore the equilibrium of the osteoarthritic joint remains a challenge. Numerous studies have revealed that dietary components and/or natural products have anti-inflammatory, antioxidant, anti-bone-resorption, and anabolic potential and have received much attention toward the development of new therapeutic strategies for OA treatment. In the present review, we provide an overview of current and emerging natural-product-based research treatments for OA management by drawing attention to experimental, pre-clinical, and clinical models. Herein, we review current and emerging natural-product-based research treatments for OA management.
Collapse
Affiliation(s)
- Maria-Luisa Pérez-Lozano
- Laboratory I3MTO, EA 4708, Université d’Orléans, CEDEX 2, 45067 Orléans, France; (M.-L.P.-L.); (A.C.); (E.L.)
- Plateforme Recherche Innovation Médicale Mutualisée d’Orléans, Centre Hospitalier Régional d’Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
| | - Annabelle Cesaro
- Laboratory I3MTO, EA 4708, Université d’Orléans, CEDEX 2, 45067 Orléans, France; (M.-L.P.-L.); (A.C.); (E.L.)
- Plateforme Recherche Innovation Médicale Mutualisée d’Orléans, Centre Hospitalier Régional d’Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
| | - Marija Mazor
- Center for Proteomics, Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia;
| | - Eric Esteve
- Service de Dermatologie, Centre Hospitalier Régional d′Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France;
| | - Sabine Berteina-Raboin
- Institut de Chimie Organique et Analytique ICOA, Université d’Orléans-Pôle de Chimie, UMR CNRS 7311, Rue de Chartres-BP 6759, CEDEX 2, 45067 Orléans, France;
| | - Thomas M. Best
- Department of Orthopedics, Division of Sports Medicine, Health Sports Medicine Institute, University of Miami, Coral Gables, FL 33146, USA;
| | - Eric Lespessailles
- Laboratory I3MTO, EA 4708, Université d’Orléans, CEDEX 2, 45067 Orléans, France; (M.-L.P.-L.); (A.C.); (E.L.)
- Plateforme Recherche Innovation Médicale Mutualisée d’Orléans, Centre Hospitalier Régional d’Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
- Centre Hospitalier Régional d’Orléans, Institut Département de Rhumatologie, 45067 Orléans, France
| | - Hechmi Toumi
- Laboratory I3MTO, EA 4708, Université d’Orléans, CEDEX 2, 45067 Orléans, France; (M.-L.P.-L.); (A.C.); (E.L.)
- Plateforme Recherche Innovation Médicale Mutualisée d’Orléans, Centre Hospitalier Régional d’Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
- Centre Hospitalier Régional d’Orléans, Institut Département de Rhumatologie, 45067 Orléans, France
| |
Collapse
|
32
|
Tang H, Hosein A, Mattioli-Belmonte M. Traditional Chinese Medicine and orthopedic biomaterials: Host of opportunities from herbal extracts. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111760. [PMID: 33545901 DOI: 10.1016/j.msec.2020.111760] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
The role of Traditional Chinese Medicine (TCM), especially herbs or herbal extracts, in treating diseases has received increasing attention. This review focuses on the use of herbal extracts as signaling molecules and functional materials in the field of orthopedics, biomaterial science and bone tissue engineering strategies. A literature review using both Chinese and English references on herbs and herbal extracts based on TCM theory used in orthopedics and biomaterial science was performed. We discuss the efficacy of herbs, the active extracts from these herbs, the combination of herbal extracts and biomaterials and, finally, the application of herbal extracts to the biomaterials specific to orthopedics. Only a few studies have confirmed the feasibility of applying herbal extracts to biomaterials to improve the role of biomaterials and/or optimize drug delivery and release in orthopedics. In this context, this review reveals a new and promising direction for herbal extracts, where the use of herbal extracts based on TCM systemic treatment, can change the limited modern medicine view of biomaterials as "only for local treatment" when considering its efficacy.
Collapse
Affiliation(s)
- Huijuan Tang
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Andrell Hosein
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
33
|
Li H, Wu R, Yu H, Zheng Q, Chen Y. Bioactive Herbal Extracts of Traditional Chinese Medicine Applied with the Biomaterials: For the Current Applications and Advances in the Musculoskeletal System. Front Pharmacol 2021; 12:778041. [PMID: 34776987 PMCID: PMC8581265 DOI: 10.3389/fphar.2021.778041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 02/05/2023] Open
Abstract
Traditional Chinese medicine (TCM) has demonstrated superior therapeutic effect for musculoskeletal diseases for thousands of years. Recently, the herbal extracts of TCM have received rapid advances in musculoskeletal tissue engineering (MTE). A literature review collecting both English and Chinese references on bioactive herbal extracts of TCM in biomaterial-based approaches was performed. This review provides an up-to-date overview of application of TCMs in the field of MTE, involving regulation of multiple signaling pathways in osteogenesis, angiogenesis, anti-inflammation, and chondrogenesis. Meanwhile, we highlight the potential advantages of TCM, opening the possibility of its extensive application in MTE. Overall, the superiority of traditional Chinese medicine turns it into an attractive candidate for coupling with advanced additive manufacturing technology.
Collapse
Affiliation(s)
- Haotao Li
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Rongjie Wu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Haiyang Yu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Qiujian Zheng, ; Yuanfeng Chen,
| | - Yuanfeng Chen
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Research Department of Medical Science, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Qiujian Zheng, ; Yuanfeng Chen,
| |
Collapse
|
34
|
Aimaiti A, Wahafu T, Keremu A, Yicheng L, Li C. Strontium Ameliorates Glucocorticoid Inhibition of Osteogenesis Via the ERK Signaling Pathway. Biol Trace Elem Res 2020; 197:591-598. [PMID: 31832923 DOI: 10.1007/s12011-019-02009-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
Glucocorticoid (GC) has been widely used in clinical work due to its anti-inflammatory and immune-inhibitory properties. However, long-term or high-dose administration is associated with side effects, such as GC-induced osteoporosis (GIOP), which causes great pain for and poses a heavy financial burden on patients. We sought to investigate the potential effects of strontium on GIOP and further explore its underlying mechanisms, including its reversal of the inhibitory effect of GC on osteogenesis of bone marrow-derived mesenchymal stem cells (BMSCs). We incubated BMSCs with Dexamethasone (DEX) in combination with or without strontium and then measured osteogenic and adipogenic gene expression levels by RT-qPCR and Western blot. We added a specific ERK signaling pathway inhibitor, U0126, to evaluate the involvement of that pathway. Strontium promoted osteogenic differentiation and matrix mineralization in DEX-treated BMSCs, accompanied by upregulation of RUNX2, Osx, ALP, BSP, COL1A1, and OCN. DEX blocked the expression of several osteogenesis-related marker genes by activating the ERK signaling pathway. U0126 attenuated the suppression of osteogenesis in DEX-treated BMSCs. These results suggested that strontium could enhance osteogenic differentiation and matrix mineralization by counteracting DEX's inhibitory effect on osteogenesis via the ERK signaling pathway. Therefore, strontium might be a promising therapeutic agent for GIOP.
Collapse
Affiliation(s)
- Abudousaimi Aimaiti
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Tuerhongjiang Wahafu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Ajimu Keremu
- Orthopedic Center, First People's Hospital of Kashgar, Kashgar, 844000, Xinjiang, China
| | - Li Yicheng
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China
| | - Cao Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, 137 South LiYuShan Road, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
35
|
Mechanisms and Molecular Targets of the Tao-Hong-Si-Wu-Tang Formula for Treatment of Osteonecrosis of Femoral Head: A Network Pharmacology Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7130105. [PMID: 32963569 PMCID: PMC7499271 DOI: 10.1155/2020/7130105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 01/11/2023]
Abstract
The Tao-Hong-Si-Wu-Tang (THSWT) formula, a classic prescription of traditional Chinese medicine, has long been used for the treatment of osteonecrosis of femoral head (ONFH). However, its mechanisms of action and molecular targets are not comprehensively clear. In the present study, the Traditional Chinese Medicine System Pharmacology (TCMSP) database was employed to retrieve the active compounds of each herb included in the THSWT formula. After identifying the drug targets of active compounds and disease targets of ONFH, intersection analysis was conducted to screen out the shared targets. The protein-protein network of the shared targets was built for further topological analysis. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis were then carried out. A gene pathway network was constructed to screen the core target genes. We identified 61 active compounds, 155 drug targets, and 5443 disease targets. However, intersection analysis only screened out 37 shared targets. Kaempferol, luteolin, and baicalein regulated the greatest number of targets associated with ONFH. The THSWT formula may regulate osteocyte function through specific biological processes, including responses to toxic substances and oxidative stress. The regulated pathways included the relaxin, focal adhesion, nuclear factor-κB, toll-like receptor, and AGE/RAGE signaling pathways. RELA, VEGFA, and STAT1 were the important target genes in the gene network associated with the THSWT formula for the treatment of ONFH. Therefore, the present study suggested that the THSWT formula has an action mechanism involving multiple compounds and network targets for the treatment of ONFH.
Collapse
|
36
|
Vimalraj S, Saravanan S, Hariprabu G, Yuvashree R, Ajieth Kanna SK, Sujoy K, Anjali D. Kaempferol-zinc(II) complex synthesis and evaluation of bone formation using zebrafish model. Life Sci 2020; 256:117993. [DOI: 10.1016/j.lfs.2020.117993] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
|
37
|
Tripathi AK, Rai D, Kothari P, Kushwaha P, Sinha S, Sardar A, Sashidhara KV, Trivedi R. Benzofuran pyran compound rescues rat and human osteoblast from lipotoxic effect of palmitate by inhibiting lipid biosynthesis and promoting stabilization of RUNX2. Toxicol In Vitro 2020; 66:104872. [DOI: 10.1016/j.tiv.2020.104872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
|
38
|
Pal S, Porwal K, Rajak S, Sinha RA, Chattopadhyay N. Selective dietary polyphenols induce differentiation of human osteoblasts by adiponectin receptor 1-mediated reprogramming of mitochondrial energy metabolism. Biomed Pharmacother 2020; 127:110207. [PMID: 32422565 DOI: 10.1016/j.biopha.2020.110207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/18/2020] [Accepted: 04/28/2020] [Indexed: 12/30/2022] Open
Abstract
Anabolic therapies for osteoporosis including dietary polyphenols promote osteoblast function by influencing its energy metabolism. Among the dietary polyphenols, the beneficial skeletal effects of genistein (an isoflavone), kaempferol (a flavone), resveratrol (RES, a stilbenoid) and epigallocatechin gallate (EGCG, a catechin) have been reported in preclinical studies. We studied the action mechanism of these nutraceuticals on osteoblast bioenergetics. All stimulated differentiation of human fetal osteoblasts (hFOB). However, only EGCG and RES stimulated mitochondrial parameters including basal and maximum respiration, spare respiratory capacity and ATP production (a measure of the activity of electron transport chain/ETC). Increases in these parameters were due to increased mitochondrial biogenesis and consequent upregulation of several mitochondrial proteins including those involved in ETC. Rotenone blocked the osteogenic effect of EGCG and RES suggesting the mediatory action of mitochondria. Both compounds rapidly activated AMPK, and dorsomorphin (an AMPK inhibitor) abolished ATP production stimulated by these compounds. Moreover, EGCG and RES upregulated the mitochondrial biogenesis factor, PGC-1α which is downstream of AMPK activation, and silencing PGC-1α blocked their stimulatory effects on ATP production and hFOB differentiation. Adiponectin receptor 1 (AdipoR1) is an upstream regulator of PGC-1α, and both compounds increased the expression of AdipoR1 but not AdipoR2. Silencing AdipoR1 blocked the upregulation of EGCG/RES-induced PGC-1α and hFOB differentiation. In rat calvarium, both compounds increased AdipoR1, PGC-1α, and RunX2 (the osteoblast transcription factor) with a concomitant increase in mitochondrial copy number and ATP levels. We conclude that EGCG and RES display osteogenic effects by reprogramming osteoblastic bioenergetics by acting as the AdipoR1 agonists.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India.
| |
Collapse
|
39
|
Dai W, Sun Y, Zhong G. A Network Pharmacology Approach to Estimate the Active Ingredients and Potential Targets of Cuscutae semen in the Treatment of Osteoporosis. Med Sci Monit 2020; 26:e920485. [PMID: 32081843 PMCID: PMC7047917 DOI: 10.12659/msm.920485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Osteoporosis is a metabolic osteopathy characterized by abnormal bone mass and microstructure that has become a public health problem worldwide. Cuscutae semen (CS) is a traditional Chinese medicine (TCM) that has a positive effect on the prevention and treatment of osteoporosis. However, the mechanism of CS is unclear. Therefore, this study aimed to reveal the possible molecular mechanism involved in the effects of CS on osteoporosis based on a network pharmacology approach. Material/Methods The inactive and active ingredients of CS were identified by searching the pharmacology analysis platform of the Chinese medicine system (TCMSP), and the targets of osteoporosis were screened in the relevant databases, such as GeneCards, PubMed, and the Comparative Toxicogenomics Database (CTD). The network of “medicine-ingredients-disease-targets (M-I-D-T)” was established by means of network pharmacology, and the key targets and core pathways were determined by R analysis. Molecular docking methods were used to evaluate the binding activity between the target and the active ingredients of CS. Results Eleven active ingredients were identified in CS, and 175 potential targets of the active ingredients were also identified from the TCMSP. Moreover, we revealed 22 539 targets related to osteoporosis in the 3 well-established databases, and we determined the intersection of the disease targets and the potential targets of the active ingredients; 107 common targets were identified and used in further analysis. Additionally, biological processes and signaling pathways involved in target action, such as fluid shear stress, atherosclerosis, cancer pathways, and the TNF signaling pathway, were determined. Finally, we chose the top 5 common targets, CCND1, EGFR, IL6, MAPK8, and VEGFA, for molecular docking with the 11 active ingredients of CS. Conclusions This study suggested that CS has multiple ingredients and multiple targets relevant to the treatment of osteoporosis. We determined that the active ingredient, sesamin, may be the most crucial ingredient of CS for the treatment of osteoporosis. Additionally, the network pharmacology method provided a novel research approach to analyze the function of complex ingredients.
Collapse
Affiliation(s)
- Weiran Dai
- Department of Cardiology Ward 1, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yue Sun
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Guoqiang Zhong
- Department of Cardiology Ward 1, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
40
|
Kaempferol protects retinal ganglion ceils from high-glucose-induced injury by regulating vasohibin-1. Neurosci Lett 2019; 716:134633. [PMID: 31743752 DOI: 10.1016/j.neulet.2019.134633] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/22/2019] [Accepted: 11/15/2019] [Indexed: 12/26/2022]
Abstract
Kaempferol is a medicinal flavonol derived from the roots of Kaempferia galanga L. Kaempferol can affect cell survival, apoptosis, and anti-oxidation, though its role and underlying mechanism in retinal ganglion cells with high-glucose injury remains unclear. In this study, we explored kaempferol's role in high-glucose injury in cells from the retinal ganglion cell (RGC) line. RGC cells were isolated and then cultured in high glucose (55 mmol/L) for 0 h, 12 h, 24 h, 48 h, or 72 h, and results showed decreased cell viability at 48 h and 72 h. We treated RGC cells with different concentrations of kaempferol (0 μmol/L, 20 μmol/L, 40 μmol/L, 60 μmol/L, 80 μmol/L, or 100 μmol/L) and high-glucose (55 mmol/L) for 48 h. The data indicated inhibited lactate dehydrogenase leakage, apoptosis, caspase-3 activity, and reactive oxygen species (ROS) levels. Moreover, whereas cell viability increased in RGC cells that were incubated with kaempferol (60 μmol/L, 80 μmol/L, or 100 μmol/L) and glucose (55 mmol/L), compared with glucose alone. Kaempferol (60 μmol/L) elevated ERK phosphorylation and vasohibin-1 (VASH1) expression, and inhibition of ERK phosphorylation reversed the effect of kaempferol (60 μmol/L) on VASH1 expression in RGC cells with high-glucose injury. Additionally, interference of VASH1 by VASH1 siRNA markedly reversed the effects of kaempferol (60 μmol/L) on cell viability, caspase-3 activity, and ROS levels in RGC cells with high glucose injury. Taken together, the results suggest that kaempferol protected retinal ganglion cells from high-glucose-induced injury via ERK and VASH1 signaling.
Collapse
|
41
|
Kim KJ, Lee Y, Son SR, Lee H, Son YJ, Lee MK, Lee M. Water Extracts of Hull-less Waxy Barley ( Hordeum vulgare L.) Cultivar 'Boseokchal' Inhibit RANKL-induced Osteoclastogenesis. Molecules 2019; 24:E3735. [PMID: 31623242 PMCID: PMC6832910 DOI: 10.3390/molecules24203735] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis is a disease that leads to reduced bone mineral density. The increase in patient and medical costs because of global aging is recognized as a problem. Decreased bone mass is a common symptom of bone diseases such as Paget's disease, rheumatoid arthritis, and multiple myeloma. Osteoclasts, which directly affect bone mass, show a marked increase in differentiation and activation in the aforementioned diseases. Moreover, these multinucleated cells made from monocytes/macrophages under the influence of RANKL and M-CSF, are the only cells capable of resorbing bones. In this study, we found that the water extracts of Boseokchal (BSC-W) inhibited osteoclast differentiation in vitro and investigated its inhibitory mechanism. BSC-W was obtained by extracting flour of Boseokchal using hexane and water. To osteoclast differentiation, bone marrow-derived macrophage cells (BMMs) were cultured with the vehicle (0.1% DMSO) or BSC-W in the presence of M-CSF and RANKL for 4 days. Cytotoxicity was measured by CCK-8. Gene expression of cells was confirmed by real-time PCR. Protein expression of cells was observed by western blot assay. Bone resorption activity of osteoclast evaluated by bone pit formation assay using an Osteo Assay Plate. BSC-W inhibited RANKL-induced osteoclastogenesis in a dose-dependent manner without exerting a cytotoxic effect on BMMs. BSC-W decreased the transcriptional and translational expression of c-Fos and NFATc1, which are regulators of osteoclastogenesis and reduced the mRNA expression level of TRAP, DC-STAMP, and cathepsin K, which are osteoclast differentiation marker. Furthermore, BSC-W reduced the resorption activity of osteoclasts. Taken together, our results indicate that BSC-W is a useful candidate for health functional foods or therapeutic agents that can help treat bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Kwang-Jin Kim
- Department of Pharmacy, Sunchon National University, Suncheon 57922, Korea.
| | - Yongjin Lee
- Department of Pharmacy, Sunchon National University, Suncheon 57922, Korea.
| | - So-Ri Son
- Department of Biomedical Science and technology, Graduate School, Kyung Hee University, Seoul 02447, Korea.
| | - Hyunjin Lee
- Department of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea.
| | - Young-Jin Son
- Department of Pharmacy, Sunchon National University, Suncheon 57922, Korea.
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Jeonnam, Suncheon 57922, Korea.
| | - Mija Lee
- Department of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea.
| |
Collapse
|
42
|
Zhao J, Wu J, Xu B, Yuan Z, Leng Y, Min J, Lan X, Luo J. Kaempferol promotes bone formation in part via the mTOR signaling pathway. Mol Med Rep 2019; 20:5197-5207. [PMID: 31638215 PMCID: PMC6854588 DOI: 10.3892/mmr.2019.10747] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 07/09/2019] [Indexed: 12/27/2022] Open
Abstract
Previous research indicates that kaempferol (Kae) promotes osteogenesis, but its underlying mechanism of action remains unclear. The present study hypothesized that the osteogenic effects of Kae were mediated through mammalian target of rapamycin (mTOR). To validate this hypothesis, bone marrow mesenchymal stem cells (BMSCs) from ovariectomized (OVX) rats were differentiated into osteoblasts. The bone mineral density and bone microarchitecture of the OVX rats was measured in vivo, while osteogenesis was evaluated in vitro via Alizarin Red S staining and alkaline phosphatase activity measurements in cultured BMSCs. The levels of phosphorylated eukaryotic translation initiation factor 4E‑binding protein 1 (p‑4E/BP1) and phosphorylated ribosomal protein S6 kinase B1 (p‑S6K), and the expression of Runt‑related transcription factor 2 and Osterix, were concurrently quantified by western blot analysis. The data suggested that Kae prevented OVX‑induced osteoporosis in rats by promoting osteoblastogenesis. Furthermore, treatment with Kae in rat BMSCs enhanced mineralization, elevated ALP activity, increased the expression levels of Runx‑2 and Osterix and increased the levels of p‑S6K and decreased the levels of p‑4E/BP1 and, consistent with its ability to promote osteoblast differentiation. In contrast, treatment with rapamycin, an mTOR inhibitor, produced the opposite phenotype. Taken together, these data suggested that the protective effects of Kae in BMSCs and in the OVX rat model resulted from the induction of osteogenesis via mTOR signaling, or at least partially via the regulation of downstream effectors of the mTOR pathway.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Jue Wu
- Laboratory of Translational Medicine, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Binwu Xu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhen Yuan
- Regeneration and Rehabilitation Engineering Research Institute on Bone and Nerve of Jiangxi, Nanchang, Jiangxi 330000, P.R. China
| | - Yu Leng
- Department of Emergency, The First People's Hospital of Jiujiang City, Jiujiang, Jiangxi 332000, P.R. China
| | - Jun Min
- Department of Rehabilitation, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Xiaoyong Lan
- Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Jun Luo
- Regeneration and Rehabilitation Engineering Research Institute on Bone and Nerve of Jiangxi, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
43
|
Wong SK, Chin KY, Ima-Nirwana S. The Osteoprotective Effects Of Kaempferol: The Evidence From In Vivo And In Vitro Studies. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3497-3514. [PMID: 31631974 PMCID: PMC6789172 DOI: 10.2147/dddt.s227738] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 09/12/2019] [Indexed: 01/06/2023]
Abstract
Kaempferol is a dietary bioflavonoid ubiquitously found in various types of plant. It possesses a wide range of medicinal properties suggesting its potential clinical utility that requires further investigation. The present review intends to highlight the efficacy of kaempferol and its molecular mechanisms of action in regulating bone metabolism. Many reports have acknowledged the bone-protecting property of kaempferol and kaempferol-containing plants using in vitro and in vivo experimental models. Kaempferol supplementation showed bone-sparing effects in newborn rats, glucocorticoid-induced and ovariectomy-induced osteoporotic models as well as bone fracture models. It achieves the bone-protective effects by inhibiting adipogenesis, inflammation, oxidative stress, osteoclastic autophagy and osteoblastic apoptosis while activating osteoblastic autophagy. The anti-osteoporotic effects of kaempferol are mediated through regulation of estrogen receptor, bone morphogenetic protein-2 (BMP-2), nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) signaling pathways. In summary, kaempferol exhibits beneficial effects on skeleton, thus is potentially effective for the prophylaxis and treatment of osteoporosis.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P, Trivedi R. FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci 2019; 229:261-276. [PMID: 31082400 DOI: 10.1016/j.lfs.2019.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
AIM Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Priyanka Kothari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
45
|
Wong MS, Poon CCW, Zhou LP, Xiao HH. Natural Products as Potential Bone Therapies. Handb Exp Pharmacol 2019; 262:499-518. [PMID: 31792676 DOI: 10.1007/164_2019_322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Demands for natural products, in the form of botanicals, dietary supplements, and herbal medicine, for management of chronic diseases are increasing globally. Natural products might be an alternative for the management of bone health to meet the demands of a growing aging population. Different types of natural products, including Chinese herbal medicine decoctions, herbs, and isolated phytochemicals, have been demonstrated to exert bone protective effects. The most common types of bone protective bioactives are flavonoids, stilbene, triterpenoids, coumestans, lignans, and phenolic acid. The actions of natural products can be mediated by acting systemically on the hormonal axis or locally via their direct or indirect effects on osteogenesis, osteoclastogenesis, as well as adipogenesis. Furthermore, with the use of metabolomic and microbiome approaches to understand the actions of natural products, novel mechanisms that involve gut-brain-bone axis are also revealed. These studies provide evidence to support the use of natural products as bone therapeutics as well as identify new biological targets for novel drug development.
Collapse
Affiliation(s)
- Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China. .,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute of The Hong Kong Polytechnic University, Shenzhen, People's Republic of China.
| | - Christina Chui-Wa Poon
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Li-Ping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Hui-Hui Xiao
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute of The Hong Kong Polytechnic University, Shenzhen, People's Republic of China
| |
Collapse
|