1
|
Wang X, Qu Z, Zhao S, Luo L, Yan L. Wnt/β-catenin signaling pathway: proteins' roles in osteoporosis and cancer diseases and the regulatory effects of natural compounds on osteoporosis. Mol Med 2024; 30:193. [PMID: 39468464 PMCID: PMC11520425 DOI: 10.1186/s10020-024-00957-x] [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: 05/17/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Osteoblasts are mainly derived from mesenchymal stem cells in the bone marrow. These stem cells can differentiate into osteoblasts, which have the functions of secreting bone matrix, promoting bone formation, and participating in bone remodeling. The abnormality of osteoblasts can cause a variety of bone-related diseases, including osteoporosis, delayed fracture healing, and skeletal deformities. In recent years, with the side effects caused by the application of PTH drugs, biphosphonate drugs, and calmodulin drugs, people have carried out more in-depth research on the mechanism of osteoblast differentiation, and are actively looking for natural compounds for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway is considered to be one of the important pathways of osteoblast differentiation, and has become an important target for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway, whether its activation is enhanced or its expression is weakened, will cause a variety of diseases including tumors. This review will summarize the effect of Wnt/β-catenin signaling pathway on osteoblast differentiation and the correlation between the related proteins in the pathway and human diseases. At the same time, the latest research progress of natural compounds targeting Wnt/β-catenin signaling pathway against osteoporosis is summarized.
Collapse
Affiliation(s)
- Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Medical University, Xi'an, China
| | - Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Luo
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Li Z, Li M, Huang S, Yu J, Liu M, Liu Y, Xu M. The expression pattern of Wnt6, Wnt10A, and HOXA13 during regenerating tails of Gekko Japonicus. Gene Expr Patterns 2024; 53:119374. [PMID: 39128795 DOI: 10.1016/j.gep.2024.119374] [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: 02/26/2024] [Revised: 04/28/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Wnt signal is crucial to correctly regenerate tissues along the original axis in many animals. Lizards are able to regenerate their tails spontaneously, while the anterior-posterior axis information required for the successful regeneration is still elusive. In this study, we investigated the expression pattern of Wnt ligands and HOX genes during regeneration. The results of in situ hybridization revealed that Wnt6 and Wnt10A mRNA levels are higher in wound epithelium (WE) than that in blastema during regeneration. In addition, we showed that Wnt agonist positively regulated the expression of HOXA13 in cultured blastema cells, while did not show similar effect on that of HOXB13, HOXC13 and HOXD13. Finally, we found that HOXA13 showed a gradient level along the anterior-posterior axis of regenerated blastema, with higher level at the caudal end. These data proposed that Wnt6, Wnt10A and HOXA13 might play an important role in establishing distal position for regeneration.
Collapse
Affiliation(s)
- Zhen Li
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Mingxuan Li
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Shuai Huang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jing Yu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Man Xu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
3
|
Chen H, Weng Z, Kalinowska M, Xiong L, Wang L, Song H, Xiao J, Wang F, Shen X. Anti-osteoporosis effect of bioactives in edible medicinal plants: a comprehensive review. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39093554 DOI: 10.1080/10408398.2024.2386449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Current treatments for osteoporosis include a calcium-rich diet, adequate exercise, and medication. Many synthetic drugs, although fast-acting, can cause a range of side effects for patients when taken over a long period, such as irritation of the digestive tract and a burden on the kidneys. As the world's population ages, the prevalence of osteoporosis is increasing, and the development of safe and effective treatments is urgently needed. Active compounds in edible and medicinal homologous plants have been used for centuries to improve bone quality. It is possible to employ them as dietary supplements to prevent osteoporosis. In this review, we analyze the influencing factors of osteoporosis and systematically summarize the research progress on the anti-osteoporosis effects of active compounds in edible and medicinal homologous plants. The literature suggests that some naturally occurring active compounds in edible and medicinal homologous plants can inhibit bone loss, prevent the degeneration of bone cell microstructure, and reduce bone fragility through alleviating oxidative stress, regulating autophagy, anti-inflammation, improving gut flora, and regulating estrogen level with little side effects. Our review provides useful guidance for the use of edible and medicinal homologous plants and the development of safer novel anti-osteoporosis dietary supplements.
Collapse
Affiliation(s)
- Huiling Chen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Zebin Weng
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jianbo Xiao
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Universidade de Vigo, Nutrition and Bromatology Group, Vigo, Spain
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
4
|
Carletti A, Gavaia PJ, Cancela ML, Laizé V. Metabolic bone disorders and the promise of marine osteoactive compounds. Cell Mol Life Sci 2023; 81:11. [PMID: 38117357 PMCID: PMC10733242 DOI: 10.1007/s00018-023-05033-x] [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: 06/20/2023] [Revised: 10/12/2023] [Accepted: 11/05/2023] [Indexed: 12/21/2023]
Abstract
Metabolic bone disorders and associated fragility fractures are major causes of disability and mortality worldwide and place an important financial burden on the global health systems. These disorders result from an unbalance between bone anabolic and resorptive processes and are characterized by different pathophysiological mechanisms. Drugs are available to treat bone metabolic pathologies, but they are either poorly effective or associated with undesired side effects that limit their use. The molecular mechanism underlying the most common metabolic bone disorders, and the availability, efficacy, and limitations of therapeutic options currently available are discussed here. A source for the unmet need of novel drugs to treat metabolic bone disorders is marine organisms, which produce natural osteoactive compounds of high pharmaceutical potential. In this review, we have inventoried the marine osteoactive compounds (MOCs) currently identified and spotted the groups of marine organisms with potential for MOC production. Finally, we briefly examine the availability of in vivo screening and validation tools for the study of MOCs.
Collapse
Affiliation(s)
- Alessio Carletti
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Paulo Jorge Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Associação Oceano Verde (GreenCoLab), Faro, Portugal
| | - Maria Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.
- Collaborative Laboratory for Sustainable and Smart Aquaculture (S2AQUAcoLAB), Olhão, Portugal.
| |
Collapse
|
5
|
Ye Z, Liu Y, Song J, Gao Y, Fang H, Hu Z, Zhang M, Liao W, Cui L, Liu Y. Expanding the therapeutic potential of Salvia miltiorrhiza: a review of its pharmacological applications in musculoskeletal diseases. Front Pharmacol 2023; 14:1276038. [PMID: 38116081 PMCID: PMC10728493 DOI: 10.3389/fphar.2023.1276038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Salvia miltiorrhiz, commonly known as "Danshen" in Chinese medicine, has longstanding history of application in cardiovascular and cerebrovascular diseases. Renowned for its diverse therapeutic properties, including promoting blood circulation, removing blood stasis, calming the mind, tonifying the blood, and benefiting the "Qi", recent studies have revealed its significant positive effects on bone metabolism. This potential has garnered attention for its promising role in treating musculoskeletal disorders. Consequently, there is a high anticipation for a comprehensive review of the potential of Salvia miltiorrhiza in the treatment of various musculoskeletal diseases, effectively introducing an established traditional Chinese medicine into a burgeoning field. AIM OF THE REVIEW Musculoskeletal diseases (MSDs) present significant challenges to healthcare systems worldwide. Previous studies have demonstrated the high efficacy and prospects of Salvia miltiorrhiza and its active ingredients for treatment of MSDs. This review aims to illuminate the newfound applications of Salvia miltiorrhiza and its active ingredients in the treatment of various MSDs, effectively bridging the gap between an established medicine and an emerging field. METHODS In this review, previous studies related to Salvia miltiorrhiza and its active ingredients on the treatment of MSD were collected, the specific active ingredients of Salvia miltiorrhiza were summarized, the effects of Salvia miltiorrhiza and its active ingredients for the treatment of MSDs, as well as their potential molecular mechanisms were reviewed and discussed. RESULTS Based on previous publications, Salvianolic acid A, salvianolic acid B, tanshinone IIA are the representative active ingredients of Salvia miltiorrhiza. Their application has shown significant beneficial outcomes in osteoporosis, fractures, and arthritis. Salvia miltiorrhiza and its active ingredients protect against MSDs by regulating different signaling pathways, including ROS, Wnt, MAPK, and NF-κB signaling. CONCLUSION Salvia miltiorrhiza and its active ingredients demonstrate promising potential for bone diseases and have been explored across a wide variety of MSDs. Further exploration of Salvia miltiorrhiza's pharmacological applications in MSDs holds great promise for advancing therapeutic interventions and improving the lives of patients suffering from these diseases.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yuyu Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yin Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang, China
| | - Haiping Fang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Zilong Hu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Wenwei Liao
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| |
Collapse
|
6
|
Arachchilage Hasitha Maduranga Karunarathne W, Hyun Choi Y, Lee MH, Kang CH, Kim GY. Gamma-aminobutyric acid (GABA)-mediated bone formation and its implications for anti-osteoporosis strategies: Exploring the relation between GABA and GABA receptors. Biochem Pharmacol 2023; 218:115888. [PMID: 38084676 DOI: 10.1016/j.bcp.2023.115888] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/18/2023]
Abstract
Osteoporosis is a significant global health concern, linked to reduced bone density and an increased fracture risk, with effective treatments still lacking. This study explored the potential of gamma-aminobutyric acid (GABA) and its receptors as a novel approach to promote osteogenesis and address osteoporosis. GABA concentrations up to 10 mM were well-tolerated by MC3T3-E1 preosteoblast, stimulating osteoblast differentiation and mineralization in a concentration- and time-dependent manner. In vivo experiments with zebrafish larvae demonstrated the ability of GABA to improve vertebral formation and enhanced bone density, indicating the potential therapeutic value for osteoporosis. Notably, GABA countered the adverse effects of prednisolone on vertebral formation, bone density, and osteogenic gene expression in zebrafish larvae, suggesting a promising therapeutic solution to counteract corticosteroid-induced osteoporosis. Moreover, our study highlighted the involvement of GABA receptors in mediating the observed osteogenic effects. By using GABAA, GABAB, and GABAC receptor antagonists, we demonstrated that blocking these receptors attenuated GABA-induced osteoblast differentiation and vertebral formation in both MC3T3-E1 cells and zebrafish larvae, underscoring the importance of GABA receptor interactions in promoting bone formation. In conclusion, these findings underscore the osteogenic potential of GABA and its ability to mitigate the detrimental effects of corticosteroids on bone health. Targeting GABA and its receptors could be a promising strategy for the development of novel therapeutic interventions to address osteoporosis. However, further investigations are warranted to fully elucidate the underlying molecular mechanism of GABA and its clinical applications in treating osteoporosis.
Collapse
Affiliation(s)
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Mi-Hwa Lee
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Chang-Hee Kang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
7
|
Moradi F, Aghamaali M, Hadavi M. Osteogenic differentiation of human amniotic mesenchymal stem cells by phycocyanin and phycoerythrin pigments isolated from Spirulina platensis and Gracilaria gracilis algae. Tissue Cell 2023; 85:102216. [PMID: 37774523 DOI: 10.1016/j.tice.2023.102216] [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: 06/14/2023] [Revised: 08/17/2023] [Accepted: 09/09/2023] [Indexed: 10/01/2023]
Abstract
Bone regeneration is a multistep and regular physiological process that occurs normally in fracture repair and bone defects. However, some factors such as aging, particular diseases and some drugs prevent or slowdown bone natural healing. Cell therapy using stem cells and differentiation activating factors is an effective treatment method for bone regeneration triggering in unusual conditions. Therefore, in the present study the effect of phycocyanin and phycoerythrin pigments which isolated from Spirulina platensis and Gracilaria gracilis algae was investigate on osteogenic differentiation potency of human Amniotic Mesenchymal Stem Cells (hAMSCs). For this purpose, hAMSCs were exposed to 300, 500, and 700 µg/ml concentrations of phycocyanin and phycoerythrin pigments and then the cells viability was measured with MTT assay in 48 and 72 h after treatment. The osteo-differentiation level of cells was studied by measuring ALP activity using calorimetric method and Alizarin red staining for calcium deposition in 7 and 21 days after treatment. Also, total RNA of cells was extracted in different time periods and then cDNA synthesized with specific primers, and relative expression of Runx2, β-catenin and Osteocalcin genes were investigated using SYBR Green RT-qPCR technique. Osteogenic differentiation of hAMSCs that treated with pigments was confirmed by mineral deposits staining and increased level of ALP activity. Furthermore, these pigments elevated significantly the expression of osteogenic marker genes compared to control samples and caused hAMSCs to differentiate into osteoblast cells. According to these results, phycocyanin and phycoerythrin may suggest as suitable osteogenic supplements with low toxicity, low cost and high efficiency, although the molecular mechanism of its efficacy is not available yet.
Collapse
Affiliation(s)
- Fatemeh Moradi
- Biochemistry Laboratory, Faculty of Science, Department of Biology, University of Guilan, Rasht, Iran
| | - Mahmoudreza Aghamaali
- Biochemistry Laboratory, Faculty of Science, Department of Biology, University of Guilan, Rasht, Iran.
| | - Mahvash Hadavi
- Tissue Engineering Laboratory, Faculty of Science, Department of Biology, University of Guilan, Rasht, Iran
| |
Collapse
|
8
|
Zhao J, Duan X, Yan S, Liu Y, Wang K, Hu M, Chai Q, Liu L, Ge C, Jia J, Dou T. Transcriptomics reveals the molecular regulation of Chinese medicine formula on improving bone quality in broiler. Poult Sci 2023; 102:103044. [PMID: 37717480 PMCID: PMC10507442 DOI: 10.1016/j.psj.2023.103044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Skeletal disorder is of concern to the poultry industry as it affects animal welfare and production performance. Traditional Chinese medicine could improve bone quality and reduce the incidence of bone disease, but the molecular regulation of Chinese medicine formula (CMF) on improving bone quality in broilers is still unclear. This study was performed to research the effects of CMF on skeletal performance of Cobb broilers and reveal the molecular regulation. A total of 120 one-day-old Cobb broilers were randomly allocated into 4 equal groups of 30 chickens, with 5 replicates and 6 chickens in each replicate. The control (CON) group was fed a diet without CMF, while the CMF1, CMF2, and CMF3 groups were supplemented with different CMF at 6,000 mg/kg diet, respectively. The broilers were raised to 60 d of age, then bone tissues were collected for biomechanical properties, micro-CT detection and transcriptomic sequencing analysis. The results showed that CMF3 improved the biomechanical properties of broiler tibia, via increasing the elastic modulus (P < 0.05), yield strength (P > 0.05), maximum stress (P < 0.05) and fracture stress (P < 0.05) of the tibia. Micro-CT analysis indicated that CMF3 increased the bone mineral density (BMD), bone volume/total volume (BV/TV), bone surface density (BS/TV), trabecular number (Tb.N), trabecular thickness (Tb.Th), and decreased the trabecular separation (Tb.Sp) of femur cancellous bone (P < 0.05). RNA-seq analysis revealed 2,177 differentially expressed genes (DEGs) (|log2FoldChange| ≥ 1, FDR < 0.05) between the CMF3 group and CON group. Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis showed 13 pathways mostly associated with bone growth and development and bone metabolism, and we identified 39 bone-related DEGs. This study suggests that CMF3 could improve bone strength and bone microstructure of broilers, and showed a positive effect on bone performance. Our research could provide a theoretical reference for the development of pollution-free feed additives to improve the skeletal performance of broilers, which could help promote healthy farming of chickens.
Collapse
Affiliation(s)
- Jingying Zhao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Xiaohua Duan
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China; Yunnan University of Chinese Medicine, 650500 Kunming, China
| | - Shixiong Yan
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Yong Liu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Kun Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Mei Hu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Qian Chai
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Lixian Liu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China; Yunnan Vocational and Technical College of Agriculture, 650031 Kunming, China
| | - Changrong Ge
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Junjing Jia
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China
| | - Tengfei Dou
- Faculty of Animal Science and Technology, Yunnan Agricultural University, 650201 Kunming, China.
| |
Collapse
|
9
|
Li J, Cao H, Zhou X, Guo J, Zheng C. Advances in the study of traditional Chinese medicine affecting bone metabolism through modulation of oxidative stress. Front Pharmacol 2023; 14:1235854. [PMID: 38027015 PMCID: PMC10646494 DOI: 10.3389/fphar.2023.1235854] [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: 06/06/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Bone metabolic homeostasis is dependent on coupled bone formation dominated by osteoblasts and bone resorption dominated by osteoclasts, which is a process of dynamic balance between bone formation and bone resorption. Notably, the formation of bone relies on the development of bone vasculature. Previous studies have shown that oxidative stress caused by disturbances in the antioxidant system of the whole organism is an important factor affecting bone metabolism. The increase in intracellular reactive oxygen species can lead to disturbances in bone metabolism, which can initiate multiple bone diseases, such as osteoporosis and osteoarthritis. Traditional Chinese medicine is considered to be an effective antioxidant. Cumulative evidence shows that the traditional Chinese medicine can alleviate oxidative stress-mediated bone metabolic disorders by modulating multiple signaling pathways, such as Nrf2/HO-1 signaling, PI3K/Akt signaling, Wnt/β-catenin signaling, NF-κB signaling, and MAPK signaling. In this paper, the potential mechanisms of traditional Chinese medicine to regulate bone me-tabolism through oxidative stress is summarized to provide direction and theoretical basis for future research related to the treatment of bone diseases with traditional Chinese medicine.
Collapse
Affiliation(s)
- Jiaying Li
- School of Sports and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Cao
- School of Sports and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xuchang Zhou
- School of Sports and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Chengqiang Zheng
- School of Sports and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Paramasivam S, Perumal SS. Methanolic extract of O.umbellata L. exhibits anti-osteoporotic effect via promoting osteoblast proliferation in MG-63 cells and inhibiting osteoclastogenesis in RANKL-stimulated RAW 264.7 cells. JOURNAL OF ETHNOPHARMACOLOGY 2023:116641. [PMID: 37236379 DOI: 10.1016/j.jep.2023.116641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/04/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Oldenlandia umbellataL., belonging to the Rubiaceae family, is an annual plant possessing anti-inflammatory and antipyretic, anti-nociceptive, anti-bacterial, anti-helminthic, antioxidant and hepatoprotective activities and used in traditional medicine to treat inflammation and respiratory diseases. AIM OF THE STUDY The present study aims to evaluate the anti-osteoporotic effect of Methanolic extract of O.umbellata in MG-63 cells and RANKL-stimulated RAW 264.7 cells. MATERIALS AND METHODS The methanolic extract from the aerial parts of O.umbellata was subjected to metabolite profiling. The anti-osteoporotic effect of MOU was assessed in MG-63 cells and RANKL-stimulated RAW 264.7 cells. In MG-63 cells, the proliferative effect of MOU was evaluated using MTT assay, ALP assay, Alizarin red staining, ELISA and western blot. Similarly, the anti-osteoclastogenic effect of MOU was assessed in RANKL-stimulated RAW 264.7 cells via MTT, TRAP staining and western blot. RESULTS LC-MS metabolite profiling showed the presence of 59 phytoconstituents including scandoside, scandoside methyl ester, deacetylasperuloside, asperulosidic acid, and cedrelopsin in MOU. In MG-63 cells, MOU has increased the proliferation of osteoblast cells and ALP activity, thereby increasing bone mineralization. ELISA results showed increased levels of osteogenic markers such as osteocalcin and osteopontin in the culture media. Western blot analysis showed inhibition of GSK3β protein expression and increased the expression levels of β-catenin, Runx-2, col 1 and osterix, promoting osteoblast differentiation. In RANKL-stimulated RAW 264.7 cells, MOU did not elicit any significant cytotoxicity; instead, it suppressed the osteoclastogenesis reducing the osteoclast number. MOU has reduced TRAP activity in a dose-dependent manner. MOU inhibited the TRAF6, NFATc1, c-Jun, C-fos and cathepsin K expression, thereby inhibiting osteoclast formation. CONCLUSION In conclusion, MOU promoted osteoblast differentiation via inhibiting GSK3β and activating Wnt/β catenin signalling and its transcription factors, including β catenin, Runx2 and Osterix. Similarly, MOU inhibited osteoclast formation by inhibiting the expression of TRAF6, NFATc1, c-Jun, C-fos and cathepsin K in RANK-RANKL signalling. Finally, it can be emphasised that O.umbellata is a potential source of therapeutic leads for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, University College of Engineering, Anna University (BIT Campus), Tiruchirappalli, 620 024, Tamil Nadu, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, University College of Engineering, Anna University (BIT Campus), Tiruchirappalli, 620 024, Tamil Nadu, India.
| |
Collapse
|
11
|
Zhong YT, Liao HB, Ye ZQ, Jiang HS, Li JX, Ke LM, Hua JY, Wei B, Wu X, Cui L. Eurycomanone stimulates bone mineralization in zebrafish larvae and promotes osteogenic differentiation of mesenchymal stem cells by upregulating AKT/GSK-3β/β-catenin signaling. J Orthop Translat 2023; 40:132-146. [PMID: 37457309 PMCID: PMC10338906 DOI: 10.1016/j.jot.2023.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/28/2023] [Accepted: 05/16/2023] [Indexed: 07/18/2023] Open
Abstract
Background Eurycomanone (EN) is a diterpenoid compound isolated from the roots of Eurycoma longifolia (E. longifolia). Previous studies have confirmed that E. longifolia can enhance bone regeneration and bone strength. We previously isolated and identified ten quassinoids from E. longifolia, and the result displayed that five aqueous extracts have the effects on promotion of bone formation, among whom EN showed the strongest activity. However, the molecular mechanism of EN on bone formation was unknown, and we further investigated in this study. Methods After the verification of purity of extracted EN, following experiments were conducted. Firstly, the pharmacologic action of EN on normal bone mineralization and the therapeutic effect of EN on Dex-induced bone loss using zebrafish larvae. The mineralization area and integral optical density (IOD) were evaluated using alizarin red staining. Then the vital signaling pathways of EN relevant to OP was identified through network pharmacology analysis. Eventually in vitro, the effect of EN on cell viability, osteogenesis activities were investigated in human bone marrow mesenchymal stem cells (hMSCs) and C3H10 cells, and the molecular mechanisms by which applying AKT inhibitor A-443654 in hMSCs. Results In zebrafish larvae, the administration in medium of EN (0.2, 1, and 5 μM) dramatically enhanced the skull mineralization area and integral optical density (IOD), and increased mRNA expressions of osteoblast formation genes (ALP, RUNX2a, SP7, OCN). Meanwhile, exposure of EN remarkably alleviated the inhibition of bone formation induced by dexamethasone (Dex), prominently improved the mineralization, up-regulated osteoblast-specific genes and down-regulated osteoclast-related genes (CTSK, RANKL, NFATc1, TRAF6) in Dex-treated bone loss zebrafish larvae. Network pharmacology outcomes showed the MAPK and PI3K-AKT signaling pathways are closely associated with 10 hub genes (especially AKT1), and AKT/GSK-3β/β-catenin was selected as the candidate analysis pathway. In hMSCs and C3H10 cells, results showed that EN at appropriate concentrations of 0.008-5 μM effectively increased the cell proliferation. In addition, EN (0.04, 0.2, and 1 μM) significantly stimulated osteogenic differentiation and mineralization as well as significantly increased the protein phosphorylation of AKT and GSK-3β, and expression of β-catenin, evidencing by the results of ALP and ARS staining, qPCR and western blotting. Whereas opposite results were presented in hMSCs when treated with AKT inhibitor A-443654, which effectively inhibited the pro-osteogenesis effect induced by EN, suggesting EN represent powerful potential in promoting osteogenesis of hMSCs, which may be closely related to the AKT/GSK-3β/β-catenin signaling pathway. Conclusions Altogether, our findings indicate that EN possesses remarkable effect on bone formation via activating AKT/GSK-3β/β-catenin signaling pathway in most tested concentrations. The translational potential of this article This study demonstrates EN is a new effective monomer in promoting bone formation, which may be a promising anabolic agent for osteoporosis (OP) treatment.
Collapse
Affiliation(s)
- Yan-ting Zhong
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hong-bo Liao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhi-qiang Ye
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Hua-sheng Jiang
- The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jia-xiao Li
- Department of Nephrology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Lin-mao Ke
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jun-ying Hua
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Bo Wei
- The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xin Wu
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, And School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
12
|
Rice Germ Ameliorated Chronic Unpredictable Mild Stress-Induced Depressive-like Behavior by Reducing Neuroinflammation. Nutrients 2022; 14:nu14245382. [PMID: 36558541 PMCID: PMC9780988 DOI: 10.3390/nu14245382] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Stress-induced neuroinflammation is widely regarded as one of the primary causes of depression. Gamma-aminobutyric acid (GABA)-enriched foods relieve stress and reduce inflammatory reactions. This study aimed to evaluate whether rice germ with 30% GABA (RG) reduced neuroinflammation in mice exposed to chronic unpredictable mild stress (CUMS). CUMS mice were administered 40, 90, and 140 mg/kg of RG. CUMS increased serum and hypothalamic pro-inflammatory cytokine (TNF-α and IL-6) levels, which were decreased by RG. In the hypothalamus, CUMS elevated M1-type microglia markers of CD86 and NF-κB, whereas RG lowered these levels. The expression levels of NLRP3 inflammasome complex (NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1), IL-1β, and IL-18 were increased in the hypothalamus of CUMS mice and decreased by RG. RG attenuated depressive-like behaviors in CUMS mice, as measured by the forced swim test and tail suspension test. In conclusion, RG decreased hypothalamic inflammation-related signals, such as TNF-α, IL-6, M1 polarization, NF-κB, NLRP3 inflammasome complex, caspase-1, IL-1β, and IL-18, to diminish depressive-like behavior.
Collapse
|
13
|
High-Intensity Low Frequency Pulsed Electromagnetic Fields Treatment Stimulates Fin Regeneration in Adult Zebrafish—A Preliminary Report. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12157768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Low-Intensity electromagnetic fields (LI-PEMFs) are known to induce a trophic stimulus on bone tissue and therefore have been largely used for the treatment of several musculoskeletal disorders. High intensity (HI) PEMFs add interesting features to bio-stimulation such as electroporation, a phenomenon characterized by transient increased cell permeabilization to molecules, and diamagnetism, a water-repulsive effect based on the diamagnetic properties of water and transmembrane ions gradients. Despite the rapid evolution of technology, the biological mechanisms underlying it are still poorly understood. In order to evaluate the effectiveness of this particular stimulation, HI LF-PEMFs were used to stimulate the caudal fin rays of adult zebrafish. Actually, the zebrafish fin regeneration is a simple, well understood, and widely adopted model for studying bone regeneration. A controlled amputation fin experiment was then conducted. Regenerated bone matrix of fin rays was dyed with calcein and then analysed under fluorescence microscopy. Both the length and the area of regenerated fin’s rays treated with HI LF-PEMFs resulted significantly increased when compared with non-treated.
Collapse
|
14
|
Combined Therapy of Yishen Zhuanggu Decoction and Caltrate D600 Alleviates Postmenopausal Osteoporosis by Targeting FoxO3a and Activating the Wnt/ β-Catenin Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7732508. [PMID: 35873637 PMCID: PMC9307327 DOI: 10.1155/2022/7732508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
Background Postmenopausal osteoporosis (PMO) is the most prevalent metabolic bone disease in women. Yishen Zhuanggu (YSZG) decoction and Caltrate D600 reportedly affects bone formation. This study aimed to investigate the efficacy and mechanism of YSZG decoction combined with Caltrate D600 in PMO treatment. Methods Ovariectomy-induced PMO rat model was treated with YSZG or/and Caltrate D600 for 12 weeks. Femur bone mineral density (BMD), osteoporosis-related protein expression, and serum parameters were measured. Pathological features of femur bone tissues were observed using hematoxylin and eosin staining. Serum levels of oxidative stress parameters were measured using corresponding commercial kits. The mRNA and protein expression of FoxO3a, Wnt, and β-catenin was detected using qRT-PCR and western blotting. Results The BMD and ultimate load of PMO rats were increased after treatment with YSZG. YSZG treatment promoted the bone trabeculae formation of PMO rats. YSZG treatment also induced bone differentiation and suppress oxidative stress in PMO rats, evidenced by the increased BALP, Runx2, OPG, SOD, and CAT levels, as well as the decreased TRACP 5b, RANKL, ROS, and MDA levels. Additionally, YSZG treatment downregulated the FoxO3a expression and upregulated the levels of Wnt and β-catenin in PMO rats. Caltrate D600 addition showed an auxiliary effect for YSZG. Conclusion YSZG decoction exerts the antiosteoporotic effect on PMO by restraining the FoxO3a expression and activating the Wnt/β-catenin pathway, which has an impressive synergistic effect with Caltrate D600.
Collapse
|
15
|
Xu L, Qian Z, Wang S, Wang R, Pu X, Yang B, Zhou Q, Du C, Chen Q, Feng Z, Xu L, Zhu Z, Qiu Y, Sun X. Galectin-3 Enhances Osteogenic Differentiation of Precursor Cells From Patients With Diffuse Idiopathic Skeletal Hyperostosis via Wnt/β-Catenin Signaling. J Bone Miner Res 2022; 37:724-739. [PMID: 35064940 DOI: 10.1002/jbmr.4508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 11/10/2022]
Abstract
Diffuse idiopathic skeletal hyperostosis (DISH) is a noninflammatory skeletal disease characterized by the progressive ectopic ossification and calcification of ligaments and enthuses. However, specific pathogenesis remains unknown. Bone marrow mesenchymal stem cells (BMSCs) are a major source of osteoblasts and play vital roles in bone metabolism and ectopic osteogenesis. However, it is unclear whether BMSCs are involved in ectopic calcification and ossification in DISH. The current study aimed to explore the osteogenic differentiation abilities of BMSCs from DISH patients (DISH-BMSCs). Our results showed that DISH-BMSCs exhibited stronger osteogenic differentiation abilities than normal control (NC)-BMSCs. Human cytokine array kit analysis showed significantly increased secretion of Galectin-3 in DISH-BMSCs. Furthermore, Galectin-3 downregulation inhibited the increased osteogenic differentiation ability of DISH-BMSCs, whereas exogenous Galectin-3 significantly enhanced the osteogenic differentiation ability of NC-BMSCs. Notably, the increased Galectin-3 in DISH-BMSCs enhanced the expression of β-catenin as well as TCF-4, whereas attenuation of Wnt/β-catenin signaling partially alleviated Galectin-3-induced osteogenic differentiation and activity in DISH-BMSCs. In addition, our results noted that Galectin-3 interacted with β-catenin and enhanced its nuclear accumulation. Further in vivo studies showed that exogenous Galectin-3 enhanced ectopic bone formation in the Achilles tendon in trauma-induced rats by activating Wnt/β-catenin signaling. The current study indicated that enhanced osteogenic differentiation of DISH-BMSCs was mainly attributed to the increased secretion of Galectin-3 by DISH-BMSCs, which enhanced β-catenin expression and its nuclear accumulation. Our study helps illuminate the mechanisms of pathological osteogenesis and sheds light on the possible development of potential therapeutic strategies for DISH treatment. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Liang Xu
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhuang Qian
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sinian Wang
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Xiaojiang Pu
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bo Yang
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingshuang Zhou
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Changzhi Du
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Quanchi Chen
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhenhua Feng
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Leilei Xu
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zezhang Zhu
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yong Qiu
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xu Sun
- Spine Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Fermented Oyster (Crassostrea gigas) Extract Cures and Prevents Prednisolone-Induced Bone Resorption by Activating Osteoblast Differentiation. Foods 2022; 11:foods11050678. [PMID: 35267311 PMCID: PMC8909515 DOI: 10.3390/foods11050678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/30/2022] Open
Abstract
Osteoporosis is a bone resorptive disease characterized by the loss of bone density, causing an increase in bone fragility. In our previous study, we demonstrated that gamma aminobutyric acid-enriched fermented oyster (Crassostrea gigas) extract (FO) stimulated osteogenesis in MC3T3-E1 preosteoblast cells and vertebral formation in zebrafish. However, the efficacy of FO in prednisolone (PDS)-induced bone resorption remains unclear. In this study, we evaluated the osteogenic potential of FO in MC3T3-E1 preosteoblast cells and zebrafish larvae under both PDS-pretreated and PDS-post-treated conditions. We found that FO recovered osteogenic activity by upregulating osteoblast markers, such as alkaline phosphatase (ALP), runt-related transcription factor 2, and osterix, in both PDS-pretreated and post-treated MC3T3-E1 osteoblast cells and zebrafish larvae. In both conditions, PDS-induced decrease in calcification and ALP activity was recovered in the presence of FO. Furthermore, vertebral resorption in zebrafish larvae induced by pretreatment and post-treatment with PDS was restored by treatment with FO, along with the recovery of osteogenic markers and downregulation of osteoclastogenic markers. Finally, whether FO disturbs the endocrine system was confirmed according to the Organization for Economic Cooperation and Development guideline 455. We found that FO did not stimulate estrogen response element-luciferase activity or proliferation in MCF7 cells. Additionally, in ovariectomized mice, no change in uterine weight was observed during FO feeding. These results indicate that FO effectively prevents and treats PDS-induced osteoporosis without endocrine disturbances.
Collapse
|
17
|
Zhang F, Attarilar S, Xie K, Han C, Huang K, Lan C, Wang C, Yang C, Wang L, Mozafari M, Li K, Liu J, Tang Y. Carfilzomib alleviated osteoporosis by targeting PSME1/2 to activate Wnt/β-catenin signaling. Mol Cell Endocrinol 2022; 540:111520. [PMID: 34838695 DOI: 10.1016/j.mce.2021.111520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Osteoporosis (OP) is characterized by decreased bone mineral density and impaired bone strength. Carfilzomib (CFZ) is a new-generation proteasome inhibitor and has been found to affect bone metabolism. However, the effect and mechanism of CFZ on OP has not been investigated systematically. In this study, we found that protein levels of proteasome activator subunit 1/2 (PSME1/2) increased in OP, and accumulated mostly in osteoblasts and osteoclasts. Treatment with PSME1/2 recombinant protein inhibited osteogenesis and promoted osteoclast formation in vitro. Also, PSME1/2 inhibited the expression of β-catenin protein, resulting in limitation of Wnt/β-catenin signaling. CFZ inhibited PSME1 and PSME2 proteasome activities and increased β-catenin protein level, resulting in the translocation of β-catenin to the nucleus and activation of canonical Wnt/β-catenin signaling, further promoting osteogenesis and inhibiting osteoclastic differentiation. In vivo, we conducted ovariectomy (OVX) to create a model of OVX-induced postmenopausal OP in mice. When analyzed by micro-CT scanning, enhancement of bone mineral density, bone volume, trabecular number, and thickness was seen in the CFZ-treated mice. Also, we noticed increased osteogenesis and decreased osteoclastogenesis, diminished expression of PSME1 and PSME2 and activated Wnt/β-catenin signaling in bone sections from OP mice treated with CFZ. Overall, our data indicated that PSME1/2 may serve as new targets for the treatment of OP, and targeting PSME1/2 with CFZ provides a candidate therapeutic molecule for postmenopausal OP.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Shokouh Attarilar
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Kegong Xie
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Chao Han
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Ke Huang
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Changgong Lan
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Chong Wang
- School of Mechanical Engineering, Dongguan University of Technology, Dongguan, Guangdong, 523808, China
| | - Chengliang Yang
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Liqiang Wang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Kai Li
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China.
| | - Jia Liu
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| | - Yujin Tang
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, 533000, Guangxi, China; Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
18
|
Kim GY, Jayasingha JA, Lee K, Choi Y, Kang CH, Lee MH. Aqueous extract of freeze-dried Protaetia brevitarsis larvae promotes osteogenesis by activating β-catenin signaling. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.338920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
19
|
Xie Z, Xu Y, Wei X, An G, Hao M, Yu Z, Qiu L. Four and a Half LIM Domains Protein 2 Mediates Bortezomib-Induced Osteogenic Differentiation of Mesenchymal Stem Cells in Multiple Myeloma Through p53 Signaling and β-Catenin Nuclear Enrichment. Front Oncol 2021; 11:729799. [PMID: 34589431 PMCID: PMC8473907 DOI: 10.3389/fonc.2021.729799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
Myeloma bone disease (MBD), caused by the inhibition of osteoblast activity and the activation of osteoclast in the bone marrow environment, is the most frequent and life-threatening complication in multiple myeloma (MM) patients. Bortezomib (Bzb) was shown to promote MM-derived mesenchymal stem cells (MM-MSCs) differentiation to osteoblast in vitro and in animal models, promoting the bone formation and regeneration, may be mediated via β-catenin/T-cell factor (TCF) pathway. Further defining molecular mechanism of Bzb-enhanced bone formation in MM will be beneficial for the treatment of myeloma patients. The present study has identified for the first time four and a half LIM domains protein 2 (FHL2), a tissue-specific coregulator that interacts with many osteogenic marker molecules, as a therapeutic target to ameliorate MM bone disease. First, increased messenger RNA (mRNA) and protein levels of FHL2, and the mRNA level of main osteoblast markers (including Runx2, ALP, and Col1A1), were found in MM-patients-derived MSCs after Bzb treatment. FHL2 KD with short hairpin RNA (shRNA) reduced the expression of osteoblast marker genes and blocked the osteogenic differentiation of MM-MSCs regardless of the presence or absence of Bzb, implying that FHL2 is an important activator of the osteogenic differentiation of human MSCs under a proteasome inhibition condition. Molecular analysis showed that the enhanced expression of FHL2 was associated with the Bzb-induced upregulation of p53. No significant change at protein level of total β-catenin was observed with or without Bzb treatment. However, it was mostly enriched to nuclei in MSCs after Bzb treatment. Moreover, β-catenin was restricted to the perinuclear region in FHL2 KD cells. These data provide evidence that FHL2 is essential for promoting β-catenin nuclear enrichment in MM-MSCs. In conclusion, FHL2 is critical for Bzb-induced osteoblast differentiation of MM-MSCs and promotes the osteogenesis, through p53 signaling and β-catenin activation. Targeting FHL2 in MM may provide a new therapeutic strategy for treating MBD.
Collapse
Affiliation(s)
- Zhenqing Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaojing Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
20
|
Karunarathne WAHM, Molagoda IMN, Lee KT, Choi YH, Jin CY, Kim GY. Anthocyanin-enriched polyphenols from Hibiscus syriacus L. (Malvaceae) exert anti-osteoporosis effects by inhibiting GSK-3β and subsequently activating β-catenin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153721. [PMID: 34461423 DOI: 10.1016/j.phymed.2021.153721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/08/2021] [Accepted: 08/15/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The bark and petal of Hibiscus syriacus L. (Malvaceae) have been used to relieve pain in traditional Korean medicine. Recently, we identified anthocyanin-enriched polyphenols from the petal of H. syriacus L. (AHs) and determined its anti-melanogenic, anti-inflammatory, and anti-oxidative properties. Nevertheless, the osteogenic potential of AHs remains unknown. PURPOSE This study was aimed to investigating the effect of AHs on osteoblast differentiation and osteogenesis in osteoblastic cell lines and zebrafish larvae. Furthermore, we investigated whether AHs ameliorates prednisolone (PDS)-induced osteoporosis. STUDY DESIGN AND METHODS Cell viability was assessed by cellular morphology, MTT assay, and flow cytometry analysis, and osteoblast differentiation was measured alizarin red staining, alkaline phosphatase (ALP) activity, and osteoblast-specific marker expression. Osteogenic and anti-osteoporotic effects of AHs were determined in zebrafish larvae. RESULTS AHs enhanced calcification and ALP activity concomitant with the increased expression of osterix (OSX), runt-related transcription factor 2 (RUNX2), and ALP in MC3T3-E1 preosteoblast and MG-63 osteosarcoma cells. Additionally, AHs accelerated vertebral formation and mineralization in zebrafish larvae, concurrent with the increased expression of OSX, RUNX2a, and ALP. Furthermore, PDS-induced loss of osteogenic activity and vertebral formation were restored by treatment with AHs, accompanied by a significant recovery of calcification, ALP activity, and osteogenic marker expression. Molecular docking studies showed that 16 components in AHs fit to glucagon synthase kinase-3β (GSK-3β); particularly, isovitexin-4'-O-glucoside most strongly binds to the peptide backbone of GSK-3β at GLY47(O), GLY47(N), and ASN361(O), with a binding score of -7.3. Subsequently, AHs phosphorylated GSK-3β at SER9 (an inactive form) and released β-catenin into the nucleus. Pretreatment with FH535, a Wnt/β-catenin inhibitor, significantly inhibited AH-induced vertebral formation in zebrafish larvae. CONCLUSION AHs stimulate osteogenic activities through the inhibition of GSK-3β and subsequent activation of β-catenin, leading to anti-osteoporosis effects.
Collapse
Affiliation(s)
| | - Ilandarage Menu Neelaka Molagoda
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung Tae Lee
- Forest Biomaterials Research Center, National Institute of Forest Science, Jinju 52817, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Cheng-Yun Jin
- Key Laboratory of Advanced Technology for Drug Preparation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
21
|
Antognelli C, Marinucci L, Frosini R, Macchioni L, Talesa VN. Metastatic Prostate Cancer Cells Secrete Methylglyoxal-Derived MG-H1 to Reprogram Human Osteoblasts into a Dedifferentiated, Malignant-like Phenotype: A Possible Novel Player in Prostate Cancer Bone Metastases. Int J Mol Sci 2021; 22:ijms221910191. [PMID: 34638532 PMCID: PMC8508123 DOI: 10.3390/ijms221910191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023] Open
Abstract
Bone metastases from prostate cancer (PCa) result from a complex cross-talk between PCa cells and osteoblasts (OB). Thus, targeting this interplay has become an attractive strategy to interfere with PCa bone dissemination. The agents currently used in clinical trials have proved ineffective, boosting research to identify additional mechanisms that may be involved in this two-directional talk. Here, we investigated whether and how 5-hydro-5-methylimidazolone (MG-H1), a specific methylglyoxal (MG)-derived advanced glycation end product (AGE), was a novel player in the dialogue between PCa and OB to drive PCa bone metastases. Conditioned medium from osteotropic PC3 PCa cells, pre-treated or not with a specific MG scavenger, was administrated to human primary OB and cell morphology, mesenchymal trans-differentiation, pro-osteogenic determinants, PCa-specific molecules, and migration/invasion were studied by phase-contrast microscopy, real-time PCR, western blot and specific assays, respectively. We found that PC3 cells were able to release MG-H1 that, by binding to the receptor for AGEs (RAGE) on OB, reprogrammed them into a less-differentiate phenotype, endowed with some PCa-specific molecular features and malignant properties, in a mechanism involving reactive oxidative species (ROS) production and NF-kB pathway activation. These findings provide novel insights into the mechanisms of PCa osteoblastic metastases and foster in vivo research toward new therapeutic strategies interfering with PCa/OB cross-talk.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (L.M.); (R.F.); (V.N.T.)
- Correspondence: ; Tel.: +39-075-585-8354
| | - Lorella Marinucci
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (L.M.); (R.F.); (V.N.T.)
| | - Roberta Frosini
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (L.M.); (R.F.); (V.N.T.)
| | - Lara Macchioni
- Department of Medicine and Surgery, Biochemistry and Physiology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy;
| | - Vincenzo Nicola Talesa
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (L.M.); (R.F.); (V.N.T.)
| |
Collapse
|
22
|
Bonnet C, Brahmbhatt A, Deng SX, Zheng JJ. Wnt signaling activation: targets and therapeutic opportunities for stem cell therapy and regenerative medicine. RSC Chem Biol 2021; 2:1144-1157. [PMID: 34458828 PMCID: PMC8341040 DOI: 10.1039/d1cb00063b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Wnt proteins are secreted morphogens that play critical roles in embryonic development, stem cell proliferation, self-renewal, tissue regeneration and remodeling in adults. While aberrant Wnt signaling contributes to diseases such as cancer, activation of Wnt/β-catenin signaling is a target of interest in stem cell therapy and regenerative medicine. Recent high throughput screenings from chemical and biological libraries, combined with improved gene expression reporter assays of Wnt/β-catenin activation together with rational drug design, led to the development of a myriad of Wnt activators, with different mechanisms of actions. Among them, Wnt mimics, antibodies targeting Wnt inhibitors, glycogen-synthase-3β inhibitors, and indirubins and other natural product derivatives are emerging modalities to treat bone, neurodegenerative, eye, and metabolic disorders, as well as prevent ageing. Nevertheless, the creation of Wnt-based therapies has been hampered by challenges in developing potent and selective Wnt activators without off-target effects, such as oncogenesis. On the other hand, to avoid these risks, their use to promote ex vivo expansion during tissue engineering is a promising application.
Collapse
Affiliation(s)
- Clémence Bonnet
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- INSERM, UMRS1138, Team 17, From Physiopathology of Ocular Diseases to Clinical Development, Paris University, Centre de Recherche des Cordeliers, and Cornea Departement, Cochin Hospital, AP-HP F-75014 Paris France
| | - Anvi Brahmbhatt
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
| | - Sophie X Deng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| | - Jie J Zheng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| |
Collapse
|
23
|
Jin S, Chang XC, Wen J, Yang J, Ao N, Zhang KY, Suo LN, Du J. Decarboxylated osteocalcin, a possible drug for type 2 diabetes, triggers glucose uptake in MG63 cells. World J Diabetes 2021; 12:1102-1115. [PMID: 34326958 PMCID: PMC8311485 DOI: 10.4239/wjd.v12.i7.1102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/05/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Uncarboxylated osteocalcin (GluOC) has been reported to improve glucose metabolism, prevent type 2 diabetes, and decrease the severity of obesity in mice with type 2 diabetes. GluOC can increase glucose uptake in a variety of cells. Glucose metabolism is the main source of energy for osteoblast proliferation and differentiation. We hypothesized that decarboxylated osteocalcin (dcOC), a kind of GluOC, can increase glucose uptake in MG63 cells (osteoblast-like osteosarcoma cells) and influence their proliferation and differentiation.
AIM To investigate the effects of dcOC on glucose uptake in human osteoblast-like osteosarcoma cells and the possible signaling pathways involved.
METHODS MG63 cells (human osteoblast-like osteosarcoma cells) were treated with dcOC (0, 0.3, 3, 10, or 30 ng/mL) for 1 and 72 h, and glucose uptake was measured by flow cytometry. The effect of dcOC on cell proliferation was measured with a CCK-8 assay, and alkaline phosphatase (ALP) enzyme activity was measured. PI3K was inhibited with LY294002, and hypoxia-inducible factor 1 alpha (HIF-1α) was silenced with siRNA. Then, GPRC6A (G protein-coupled receptor family C group 6 subtype A), total Akt, phosphorylated Akt, HIF-1α, and glucose transporter 1 (GLUT1) levels were measured by Western blot to elucidate the possible pathways by which dcOC modulates glucose uptake.
RESULTS The glucose uptake of MG63 cells was significantly increased compared with that of the paired control cells after short-term (1 h) treatment with dcOC at different concentrations (0.3, 3, and 10 ng/mL groups, P < 0.01; 30 ng/mL group, P < 0.05). Glucose uptake of MG63 cells was significantly increased compared with that of the paired control cells after long-term (72 h) treatment with dcOC at different concentrations (0.3, 3, and 10 ng/mL groups, P < 0.01; 30 ng/mL group, P < 0.05). DcOC triggered Akt phosphorylation in a dose-dependent manner, and the most effective stimulatory concentration of dcOC for short-term (1 h) was 3 ng/mL (P < 0.01). LY294002 abolished the dcOC-mediated (1 h) promotion of Akt phosphorylation and glucose uptake without affecting GLUT1 protein expression. Long-term dcOC stimulation triggered Akt phosphorylation and increased the protein levels of HIF-1α, GLUT1, and Runx2 in a dose-dependent manner. Inhibition of HIF-1α with siRNA abolished the dcOC-mediated glucose uptake and substantially decreased GLUT1 protein expression. DcOC intervention promoted cell proliferation in a time- and dose-dependent manner as determined by the CCK-8 assay. Treatment with both 3 ng/mL and 10 ng/mL dcOC affected the ALP activity in MG63 cells after 72 h (P < 0.01).
CONCLUSION Short- and long-term dcOC treatment can increase glucose uptake and affect proliferation and ALP activity in MG63 cells. This effect may occur through the PI3K/Akt, HIF-1α, and GLUT1 signaling factors.
Collapse
Affiliation(s)
- Shi Jin
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Xiao-Cen Chang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Jing Wen
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Jing Yang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Na Ao
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Ke-Ying Zhang
- Department of Endocrinology, The Fifth People’s Hospital of Shenyang, Shenyang 110023, Liaoning Province, China
| | - Lin-Na Suo
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Jian Du
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| |
Collapse
|
24
|
Molagoda IMN, Kang CH, Lee MH, Choi YH, Lee CM, Lee S, Kim GY. Fisetin promotes osteoblast differentiation and osteogenesis through GSK-3β phosphorylation at Ser9 and consequent β-catenin activation, inhibiting osteoporosis. Biochem Pharmacol 2021; 192:114676. [PMID: 34256044 DOI: 10.1016/j.bcp.2021.114676] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
Fisetin is a bioactive flavonol that inhibits osteoclastogenesis and promotes osteoblastogenesis. However, the osteogenic activity of fisetin needs to be comprehensively elucidated. In the present study, we observed that fisetin significantly increased alkaline phosphatase (ALP) activity and bone mineralization in MC3T3-E1 preosteoblasts, accompanied by a significant increase in runt-related transcription factor 2 (RUNX2), ALP, collagen type Ⅰ alpha 1 (Col1α1), osterix (OSX), osteocalcin (OCN), and bone morphogenetic protein 4 (BMP4) expression. Furthermore, fisetin promoted vertebral formation in zebrafish larvae, with the highest fisetin concentration comparable with that observed in β-glycerophosphate treatment. Fisetin also inhibited prednisolone (PDS)-induced anti-osteoblastic genes, including nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), tartrate-resistant acid phosphatase-6 (ACP6), dendritic cell-specific transmembrane protein (DC-STAMP), and cathepsin K (CTSK). Fisetin potently mitigated the PDS-induced inhibition of ALP activity and bone mineralization, as well as vertebral resorption in zebrafish larvae. Moreover, we confirmed that fisetin-induced osteogenic effect was activated through phosphorylation of glycogen synthase kinase-3β (GSK-3β) at Ser9, consequently releasing β-catenin from the destructive complex to promote its nuclear translocation. β-Catenin inhibition by FH535 and the stabilization of GSK-3β by DOI hydrochloride remarkably inhibited fisetin-induced osteogenic activities, indicating that the GSK-3β/β-catenin signaling pathway plays a vital role in fisetin-induced osteogenesis. Collectively, our findings suggest that fisetin stimulates osteogenic activity and could be used as an effective strategy to prevent bone resorption.
Collapse
Affiliation(s)
- Ilandarage Menu Neelaka Molagoda
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Chang-Hee Kang
- Nakdongggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Mi-Hwa Lee
- Nakdongggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, RI 02912, USA
| | - Seungheon Lee
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
25
|
Hydroxysafflor yellow A promotes osteogenesis and bone development via epigenetically regulating β-catenin and prevents ovariectomy-induced bone loss. Int J Biochem Cell Biol 2021; 137:106033. [PMID: 34216755 DOI: 10.1016/j.biocel.2021.106033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
In clinical treatment, there is increasingly prevalent that traditional Chinese medicine treats common bone diseases including osteoporosis. Hydroxysafflor yellow A (HSYA), one of the essential compounds of Safflower, has been used as the therapy for thrombus, myocardial ischemia, and inflammation, but its effect on osteogenesis through epigenetic control and ovariectomy-induced bone loss in vivo has not been explored. Therefore, the study aimed to explore the function and mechanism of HSYA on bone formation and development. We found HSYA could enhance the cell viability and promote osteogenesis of hBMSCs in vitro. Mechanistically, HSYA could increase the expression of β-catenin leading to its accumulation in the nucleus and activation of downstream targets to promote osteogenesis. Besides, RNA-seq and quantitative RT-PCR and western blot showed KDM7A was significantly increased by HSYA. The occupancy of H3K27me2 on β-catenin promoter was significantly decreased by HSYA, which could be reversed by silencing endogenous KDM7A. More importantly, HSYA promoted bone development in chick embryos and prevented ovariectomy (OVX)-induced bone loss in SD rats. Taken together, our study has shown convincing evidence that HSYA could promote osteogenesis and bone development via epigenetically regulating β-catenin and prevent ovariectomy-induced bone loss.
Collapse
|
26
|
Zhao Z, Wang C, Xu Y, Wang X, Jia B, Yu T, Wang Y, Zhang Y. Effects of the Local Bone Renin-Angiotensin System on Titanium-Particle-Induced Periprosthetic Osteolysis. Front Pharmacol 2021; 12:684375. [PMID: 34248634 PMCID: PMC8264785 DOI: 10.3389/fphar.2021.684375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/08/2021] [Indexed: 12/28/2022] Open
Abstract
Wear particles may induce osteoclast formation and osteoblast inhibition that lead to periprosthetic osteolysis (PPOL) and subsequent aseptic loosening, which is the primary reason for total joint arthroplasty failure. Local bone renin-angiotensin system (RAS) has been found to participate in the pathogenic process of various bone-related diseases via promoting bone resorption and inhibiting bone formation. However, it remains unclear whether and how local bone RAS participates in wear-particle-induced PPOL. In this study, we investigated the potential role of RAS in titanium (Ti) particle-induced osteolysis in vivo and osteoclast and osteoblast differentiation in vitro. We found that the expressions of AT1R, AT2R and ACE in the interface membrane from patients with PPOL and in calvarial tissues from a murine model of Ti-particle-induced osteolysis were up-regulated, but the increase of ACE in the calvarial tissues was abrogated by perindopril. Moreover, perindopril mitigated the Ti-particle-induced osteolysis in the murine model by suppressing bone resorption and increasing bone formation. We also observed in RAW264.7 macrophages that Ang II promoted but perindopril suppressed Ti-particle-induced osteoclastogenesis, osteoclast-mediated bone resorption and expression of osteoclast-related genes. Meanwhile, Ang II enhanced but perindopril repressed Ti-particle-induced suppression of osteogenic differentiation and expression of osteoblast-specific genes in mouse bone marrow mesenchymal stem cells (BMSCs). In addition, local bone RAS promoted Ti-particle-induced osteolysis by increasing bone resorption and decreasing bone formation through modulating the RANKL/RANK and Wnt/β-catenin pathways. Taken together, we suggest that inhibition of RAS may be a potential approach to the treatment of wear-particle-induced PPOL.
Collapse
Affiliation(s)
- Zhiping Zhao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China.,Medical Department of Qingdao University, Qingdao, China
| | - Changyao Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingxing Xu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China.,Medical Department of Qingdao University, Qingdao, China
| | - Xiangyu Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China.,Medical Department of Qingdao University, Qingdao, China
| | - Bin Jia
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China.,Medical Department of Qingdao University, Qingdao, China
| | - Tengbo Yu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingzhen Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongtao Zhang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Xie B, Chen S, Xu Y, Han W, Hu R, Chen M, Zhang Y, Ding S. The Impact of Glucagon-Like Peptide 1 Receptor Agonists on Bone Metabolism and Its Possible Mechanisms in Osteoporosis Treatment. Front Pharmacol 2021; 12:697442. [PMID: 34220521 PMCID: PMC8243369 DOI: 10.3389/fphar.2021.697442] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus and osteoporosis are closely related and have complex influencing factors. The impact of anti-diabetic drugs on bone metabolism has received more and more attention. Type 2 diabetes mellitus (T2DM) would lead to bone fragility, high risk of fracture, poor bone repair and other bone-related diseases. Furthermore, hypoglycemic drugs used to treat T2DM may have notable detrimental effects on bones. Thus, the clinically therapeutic strategy for T2DM should not only effectively control the patient's glucose levels, but also minimize the complications of bone metabolism diseases. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are novel and promising drug for the treatment of T2DM. Some studies have found that GLP-1RAs may play an anti-osteoporotic effect by controlling blood sugar levels, promoting bone formation and inhibiting bone resorption. However, in clinical practice, the specific effects of GLP-1RA on fracture risk and osteoporosis have not been clearly defined and evidenced. This review summarizes the current research findings by which GLP-1RAs treatment of diabetic osteoporosis, postmenopausal osteoporosis and glucocorticoid-induced osteoporosis and describes possible mechanisms, such as GLP-1R/MAPK signaling pathway, GLP-1R/PI3K/AKT signaling pathway and Wnt/β-catenin pathway, that are associated with GLP-1RAs and osteoporosis. The specific role and related mechanisms of GLP-1RAs in the bone metabolism of patients with different types of osteoporosis need to be further explored and clarified.
Collapse
Affiliation(s)
- Baocheng Xie
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Shichun Chen
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yongxiang Xu
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Weichao Han
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Runkai Hu
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Minyi Chen
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yusheng Zhang
- Department of Pharmacy, The First People's Hospital of Foshan (The Affiliated Foshan Hospital of Sun Yat-Sen University), Foshan, China
| | - Shaobo Ding
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| |
Collapse
|
28
|
Lin FJ, Li H, Wu DT, Zhuang QG, Li HB, Geng F, Gan RY. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit Rev Food Sci Nutr 2021; 62:8646-8674. [PMID: 34058920 DOI: 10.1080/10408398.2021.1931023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish is a species of freshwater fish, popular in aquariums and laboratories. Several advantageous features have facilitated zebrafish to be extensively utilized as a valuable vertebrate model in the lab. It has been well-recognized that natural products possess multiple health benefits for humans. With the increasing demand for natural products in the development of functional foods, nutraceuticals, and natural cosmetics, the zebrafish has emerged as an unprecedented tool for rapidly and economically screening and identifying safe and effective substances from natural products. This review first summarized the key factors for the management of zebrafish in the laboratory, followed by highlighting the current progress on the establishment and applications of zebrafish models in the bioactivity evaluation of natural products. In addition, the zebrafish models used for assessing the potential toxicity or health risks of natural products were involved as well. Overall, this review indicates that zebrafish are promising animal models for the bioactivity and safety evaluation of natural products, and zebrafish models can accelerate the discovery of novel natural products with potential health functions.
Collapse
Affiliation(s)
- Fang-Jun Lin
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Hang Li
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Qi-Guo Zhuang
- China-New Zealand Belt and Road Joint Laboratory on Kiwifruit, Sichuan Provincial Academy of Natural Resource Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Ren-You Gan
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
29
|
Hong J, Ye F, Yu B, Gao J, Qi F, Wang W. Identification of the specific microRNAs and competitive endogenous RNA mechanisms in osteoporosis. J Int Med Res 2021; 48:300060520954722. [PMID: 33021861 PMCID: PMC7543140 DOI: 10.1177/0300060520954722] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective Osteoporosis and osteoarthritis are metabolic skeletal disorders. This study
aimed to identify specific networks of competitive endogenous RNA (ceRNA) in
osteoporosis that differ from those in osteoarthritis. Methods The dataset GSE74209 was downloaded from the Gene Expression Omnibus, and
differentially expressed microRNAs (DEmiRNAs) in osteoporotic samples and
osteoarthritic samples were identified. After predicting target genes and
linked long noncoding (lnc)RNAs, ceRNA networks of DEmiRNAs were
constructed. The nodes that overlapped between ceRNA networks and the
Comparative Toxicogenomics Database were selected as key candidates. Results Fifteen DEmiRNAs (including 2 downregulated and 13 upregulated miRNAs) were
identified in osteoporotic samples versus osteoarthritic samples; these
targeted 161 genes and linked to 60 lncRNAs. The ceRNA network consisted of
6 DEmiRNAs, 63 target genes, and 53 lncRNAs. After searching the Comparative
Toxicogenomics Database and mining the literature, 2 lncRNAs
(MALAT1 and NEAT1), 2 DEmiRNAs
(hsa-miR-32-3p,
downregulated; and hsa-miR-22-3p, upregulated) and 6 genes
(SP1, PTEN, ESR1,
ERBB3, CSF1R, and
CDK6) that relate to cell death, growth, and
differentiation were identified as key candidates separating osteoporosis
from osteoarthritis. Conclusions Two miRNA–ceRNA networks (including
NEAT1/MALAT1-hsa-miR-32-3p-SP1/FZD6
and
NEAT1/MALAT1-hsa-miR-22-3p-PTEN/ESR1/ERBB3/CSF1R/CDK6)
might have crucial and specific roles in osteoporosis.
Collapse
Affiliation(s)
- Junyi Hong
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| | - Fusheng Ye
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| | - Binjia Yu
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| | - Junwei Gao
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| | - Feicheng Qi
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| | - Wei Wang
- Department of Orthopaedics, Zhejiang Xiaoshan Hospital, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
30
|
Li R, Dong Y, Li F. ETS Proto-Oncogene 1 Suppresses MicroRNA-128 Transcription to Promote Osteogenic Differentiation Through the HOXA13/β-Catenin Axis. Front Physiol 2021; 12:626248. [PMID: 33746773 PMCID: PMC7965964 DOI: 10.3389/fphys.2021.626248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
ETS proto-oncogene 1 (ETS1) has been implicated in osteoporosis (OP), but the exact molecular mechanisms are complex. This work focuses on the impact of ETS1 on the osteogenic differentiation and the molecules involved. A mouse pre-osteoblast cell line MC3T3-E1 was used for in vitro experiments. ETS1 was upregulated during the process of osteogenic differentiation of MC3T3-E1 cells. Overexpression of ETS1 promoted expression of osteogenic markers, alkaline phosphate concentration, and calcareous accumulation in cells. ETS1 was found to specifically bind to miR-128 promoter to suppress its transcription, while miR-128 could target homeobox A13 (HOXA13). Therefore, ETS1 suppressed miR-128 transcription to upregulate HOXA13 expression. Overexpression of HOXA13 promoted the osteogenic differentiation ability of cells and increased the protein level of β-catenin. Either overexpression of miR-128 or downregulation of β-catenin by CWP232228, a β-catenin-specific antagonist, blocked the promoting roles of ETS1 in cells. To conclude, this study provided evidence that ETS1 suppresses miR-128 transcription to activate the following HOXA13/β-catenin axis, therefore promoting osteogenic differentiation ability of MC3T3-E1 cells. This finding may offer novel ideas for OP treatment.
Collapse
Affiliation(s)
- Renyao Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Ying Dong
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Feipeng Li
- Naton Biotech (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
31
|
Gao L, Gou N, Yao M, Amakye WK, Ren J. Food-derived natural compounds in the management of chronic diseases via Wnt signaling pathway. Crit Rev Food Sci Nutr 2021; 62:4769-4799. [PMID: 33554630 DOI: 10.1080/10408398.2021.1879001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Wnt signaling pathway is an evolutionarily conserved pathway that control embryonic development, adult tissue homeostasis, and pathological processes of organisms throughout life. However, dysregulation of the Wnt signaling is associated with the occurrence of chronic diseases. In comparison with the application of chemical drugs as traditional treatment for chronic diseases, dietary agents have unique advantages, such as less side effects, multiple targets, convenience in accessibility and higher acceptability in long-term intervention. In this review, we summarized current progress in manipulating the Wnt signaling using food components and its benefits in managing chronic diseases. The underlying mechanisms of bioactive food components in the management of the disease progression via the Wnt signaling was illustrated. Then, the review focused on the function of dietary pattern (which might act via combination of foods with multiple nutrients or food ingredients) on targeting Wnt signaling at multiple level. The potential caveats and challenges in developing new strategy via modulating Wnt-associated diseases with food-based agents and appropriate dietary pattern are also discussed in detail. This review shed light on the understanding of the regulatory effect of food bioactive components on chronic diseases management through the Wnt signaling, which can be expanded to other specific signaling pathway associated with disease.
Collapse
Affiliation(s)
- Li Gao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Na Gou
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Maojin Yao
- Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - William Kwame Amakye
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Jiaoyan Ren
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China.,Research Institute for Food Nutrition and Human Health, Guangzhou, China
| |
Collapse
|
32
|
Jeong A, Park BC, Kim HY, Choi JY, Cheon J, Park JH, Lee BJ, Kim K. Efficacy and safety of fermented oyster extract for height of children with short stature: a randomized placebo-controlled trial. Integr Med Res 2020; 10:100691. [PMID: 33680842 PMCID: PMC7918253 DOI: 10.1016/j.imr.2020.100691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Background Some experimental studies have established the effect of oysters on the promotion of body growth. Yet, there is a lack of human clinical studies. The objective of this study was to evaluate the effect of a fermented oyster (FO) extract on the increase in the height of children with stature in the 25th percentile by age. Methods In total, 100 children (6–11 years old) were randomly divided into two (FO or control) groups. For 24 weeks, the subjects in the FO group took the FO extract once daily before sleeping, whereas the control group took placebo extracts, simultaneously. We evaluated the height gain, height velocity (HV), height standard deviation score (SDS), urine deoxypyridinoline (DPD), growth hormone (GH), insulin-like growth factor (IGF-1), and IGF binding protein 3 (IGFBP-3). Results The height gain and height SDS were significantly higher in the FO group than in the placebo group after 24 weeks (height gain: p < 0.001, height SDS: p < 0.005). The HV was also significantly higher in the FO group than in the placebo group after the 6th and 24th week (p = 0.001, p = 0.004). After 24 weeks, we observed a decrease in GH, IGF, and IGFBP-3 in both groups. However, serum IGFBP-3 level in the FO group reduced less than placebo group. Conclusion FO supplementation may help to increase the height of children, and the effect might be mediated via effects on the IGFBP-3 levels.
Collapse
Affiliation(s)
- Aram Jeong
- Department of Korean Pediatrics, School of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Beom-Chan Park
- Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hee-Yeon Kim
- Department of Korean Pediatrics, Korean Medicine Hospital, Pusan National University, Yangsan, Republic of Korea
| | - Jun-Yong Choi
- Department of Internal Medicine, School of Korean Medicine, and Korean Medicine Hospital of Pusan National University, Yangsan, Republic of Korea
| | - Jinhong Cheon
- Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea.,Department of Korean Pediatrics, Korean Medicine Hospital, Pusan National University, Yangsan, Republic of Korea
| | | | - Bae-Jin Lee
- Marine Bioprocess Co. Ltd., Busan, Republic of Korea
| | - Kibong Kim
- Department of Korean Pediatrics, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea.,Department of Korean Pediatrics, Korean Medicine Hospital, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
33
|
Lee H, Hwangbo H, Ji SY, Kim MY, Kim SY, Kim DH, Hong SH, Lee SJ, Assefa F, Kim GY, Park EK, Park JH, Lee BJ, Jeon YJ, Choi YH. Gamma Aminobutyric Acid-Enriched Fermented Oyster ( Crassostrea gigas) Increases the Length of the Growth Plate on the Proximal Tibia Bone in Sprague-Dawley Rats. Molecules 2020; 25:molecules25194375. [PMID: 32977643 PMCID: PMC7582314 DOI: 10.3390/molecules25194375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 11/16/2022] Open
Abstract
Bone growth during childhood and puberty determines an adult’s final stature. Although several prior studies have reported that fermented oyster (FO) consisting of a high amount of gamma aminobutyric acid can be attributed to bone health, there is no research on the efficacy of FO on growth regulation and the proximal tibial growth plate. Therefore, in this study, we investigated the effect of FO oral administration on hepatic and serum growth regulator levels and the development of the proximal tibial growth plate in young Sprague-Dawley rats. Both oral administration of FO (FO 100, 100 mg/kg FO and FO 200, 200 mg/kg FO) and subcutaneous injection of recombinant human growth hormone (rhGH, 200 μg/kg of rhGH) for two weeks showed no toxicity. Circulating levels of growth hormone (GH) significantly increased in the FO 200 group. The expression and secretion of insulin-like growth factor-1 (IGF-1) and insulin-like growth factor binding protein-3 (IGFBP-3) were enhanced by FO administration. FO administration promoted the expression of bone morphogenic proteins IGF-1 and IGFBP-3 in the proximal tibial growth plate. This positive effect of FO resulted in incremental growth of the entire plate length by expanding the proliferating and hypertrophic zones in the proximal tibial growth plate. Collectively, our results suggested that oral administration of FO is beneficial for bone health, which may ultimately result in increased height.
Collapse
Affiliation(s)
- Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Smart Bio-Health, Dong-eui University, Busan 47340, Korea
| | - Su Hyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Su Jeong Lee
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Freshet Assefa
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (G.-Y.K.); (Y.-J.J.)
| | - Eui Kyun Park
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Joung-Hyun Park
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan 46048, Korea; (J.-H.P.); (B.-J.L.)
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan 46048, Korea; (J.-H.P.); (B.-J.L.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (G.-Y.K.); (Y.-J.J.)
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
- Correspondence: ; Tel.: +82-51-890-3319
| |
Collapse
|
34
|
Fermented Oyster Extract Promotes Insulin-Like Growth Factor-1-Mediated Osteogenesis and Growth Rate. Mar Drugs 2020; 18:md18090472. [PMID: 32962034 PMCID: PMC7551862 DOI: 10.3390/md18090472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Fermented oyster (Crassostrea gigas) extract (FO) prevents ovariectomy-induced osteoporosis by inhibiting osteoclastogenesis and activating osteogenesis. However, the molecular mechanisms underlying FO-mediated bone formation and growth rate are unclear. In the current study, we found that FO significantly upregulated the expression of growth-promoting genes in zebrafish larvae including insulin-like growth factor 1 (zigf-1), insulin-like growth factor binding protein 3 (zigfbp-3), growth hormone-1 (zgh-1), growth hormone receptor-1 (zghr-1), growth hormone receptor alpha (zghra), glucokinase (zgck), and cholecystokinin (zccka). In addition, zebrafish larvae treated with 100 μg/mL FO increased in total body length (3.89 ± 0.13 mm) at 12 days post fertilization (dpf) compared to untreated larvae (3.69 ± 0.02 mm); this effect was comparable to that of the β-glycerophosphate-treated zebrafish larvae (4.00 ± 0.02 mm). Furthermore, FO time- and dose-dependently increased the extracellular release of IGF-1 from preosteoblast MC3T3-E1 cells, which was accompanied by high expression of IGF-1. Pharmacological inhibition of IGF-1 receptor (IGF-1R) using picropodophyllin (PPP) significantly reduced FO-mediated vertebrae formation (from 9.19 ± 0.31 to 5.53 ± 0.35) and growth performance (from 3.91 ± 0.02 to 3.69 ± 0.01 mm) in zebrafish larvae at 9 dpf. Similarly, PPP significantly decreased FO-induced calcium deposition in MC3T3-E1 cells by inhibiting GSK-3β phosphorylation at Ser9. Additionally, DOI hydrochloride, a potent stabilizer of GSK-3β, reduced FO-induced nuclear translocation of RUNX2. Transient knockdown of IGF-1Rα/β using specific silencing RNA also resulted in a significant decrease in calcium deposition and reduction in GSK-3β phosphorylation at Ser9 in MC3T3-E1 cells. Altogether, these results indicate that FO increased phosphorylated GSK-3β at Ser9 by activating the autocrine IGF-1-mediated IGF-1R signaling pathway, thereby promoting osteogenesis and growth performance. Therefore, FO is a potential nutritional supplement for bone formation and growth.
Collapse
|
35
|
Park C, Lee H, Han MH, Jeong JW, Kim SO, Jeong SJ, Lee BJ, Kim GY, Park EK, Jeon YJ, Choi YH. Cytoprotective effects of fermented oyster extracts against oxidative stress-induced DNA damage and apoptosis through activation of the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts. EXCLI JOURNAL 2020; 19:1102-1119. [PMID: 33013267 PMCID: PMC7527492 DOI: 10.17179/excli2020-2376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
Abstract
Osteoblast damage by oxidative stress has been recognized as a cause of bone-related disease, including osteoporosis. Recently, we reported that fermented Pacific oyster (Crassostrea gigas) extracts (FO) inhibited osteoclastogenesis and osteoporosis, while promoting osteogenesis. However, since the beneficial potential of FO on osteoblasts is not well known, in the present study, we investigated the cytoprotective effect of FO against oxidative stress in MC3T3-E1 osteoblasts. Our results demonstrated that FO inhibited hydrogen peroxide (H2O2)-induced DNA damage and cytotoxicity through the rescue of mitochondrial function by blocking abnormal ROS accumulation. FO also prevented apoptosis by suppressing loss of mitochondrial membrane potential and cytosolic release of cytochrome c, decreasing the rate of Bax/Bcl-2 expression and reducing the activity of caspase-9 and caspase-3 in H2O2-stimulated MC3T3-E1 osteoblasts, suggesting that FO protected MC3T3-E1 osteoblasts from the induction of caspase dependent- and mitochondria-mediated apoptosis by oxidative stress. In addition, FO markedly promoted the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was associated with the enhanced expression of heme oxygenase-1 (HO-1). However, inhibiting the expression of HO-1 by artificially blocking the expression of Nrf2 using siRNA significantly eliminated the protective effect of FO, indicating that FO activates the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts to protect against oxidative stress. Based on the present data, FO is thought to be useful as a potential therapeutic agent for the inhibition of oxidative stress in osteoblasts.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong?eui University, Busan, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Min Ho Han
- National Marine Biodiversity Institute of Korea, Seocheon, Republic of Korea
| | - Jin-Woo Jeong
- Freshwater Bioresources Utilization Bureau, Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Sung Ok Kim
- Department of Food Science and Biotechnology, College of Engineering, Kyungsung University, Busan, Republic of Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Science, Youngsan University, Yangsan, Republic of Korea
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| |
Collapse
|
36
|
Li K, Zhuang P, Tao B, Li D, Xing X, Mei X. Ultra-Small Lysozyme-Protected Gold Nanoclusters as Nanomedicines Inducing Osteogenic Differentiation. Int J Nanomedicine 2020; 15:4705-4716. [PMID: 32636626 PMCID: PMC7335297 DOI: 10.2147/ijn.s241163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Ultra-small gold nanoclusters (AuNCs), as emerging fluorescent nanomaterials with excellent biocompatibility, have been widely investigated for in vivo biomedical applications. However, their effects in guiding osteogenic differentiation have not been investigated, which are important for osteoporosis therapy and bone regeneration. Herein, for the first time, lysozyme-protected AuNCs (Lys-AuNCs) are used to stimulate osteogenic differentiation, which have the potential for the treatment of bone disease. METHODS Proliferation of MC3T3E-1 is important for osteogenic differentiation. First, the proliferation rate of MC3T3E-1 was studied by Cell Counting Kit-8 (CCK8) assays. Signaling pathways of PI3K/Akt play central roles in controlling proliferation throughout the body. The expression of PI3K/Akt was investigated in the presence of lysozyme, and lysozyme-protected AuNCs (Lys-AuNCs) by Western blot (WB) and intracellular cell imaging to evacuate the osteogenic differentiation mechanisms. Moreover, the formation of osteoclasts (OC) plays a negative role in the differentiation of osteoblasts. Nuclear factor κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) signaling pathways are used to understand the negative influence of the osteogenic differentiation by the investigation of Raw 264.7 cell line. Raw 264.7 (murine macrophage-like) cells and NIH/3T3 (mouse fibroblast) cells were treated with tyloxapol, and the cell viability was assessed. Raw 264.7 cells have also been used for in vitro studies, on understanding the osteoclast formation and function. The induced osteoclasts were identified by TRAP confocal fluorescence imaging. These key factors in osteoclast formation, such as (NFATc-1, c-Fos, V-ATPase-2 and CTSK), were investigated by Western blot. RESULTS Based on the above investigation, Lys-AuNCs were found to promote osteogenic differentiation and decrease osteoclast activity. It is noteworthy that the lysozyme (protected template), AuNPs, or the mixture of Lysozyme and AuNPs have negligible effects on osteoblastic differentiation compared to Lys-AuNCs. CONCLUSION This study opens up a novel avenue to develop a new gold nanomaterial for promoting osteogenic differentiation. The possibility of using AuNCs as nanomedicines for the treatment of osteoporosis can be expected.
Collapse
Affiliation(s)
- Kuo Li
- Department of Orthopedics, School of Pharmaceutical Science, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Pengfei Zhuang
- Department of Orthopedics, School of Pharmaceutical Science, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
- Department of Basic Science, School of Pharmaceutical Science, Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Bailong Tao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing400044, People’s Republic of China
| | - Dan Li
- Department of Orthopedics, School of Pharmaceutical Science, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
- Department of Basic Science, School of Pharmaceutical Science, Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Xuejiao Xing
- Department of Orthopedics, School of Pharmaceutical Science, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Xifan Mei
- Department of Orthopedics, School of Pharmaceutical Science, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| |
Collapse
|
37
|
Lee H, Hwang-Bo H, Ji SY, Kim MY, Kim SY, Woo M, Keum YS, Noh JS, Park JH, Lee BJ, Kim GY, Park EK, Chang YC, Jeon YJ, Choi YH. Effect of fermented oyster extract on growth promotion in Sprague-Dawley rats. Integr Med Res 2020; 9:100412. [PMID: 32509520 PMCID: PMC7264051 DOI: 10.1016/j.imr.2020.100412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Background Oysters (Crassostrea gigas) are a popular marine product worldwide and have the advantage of nutritional benefits. This study aimed to investigate the effect of fermented oyster extract (FO) on growth promotion, including analysis of body size, bone microarchitecture, hematology and biochemistry in vivo. Methods The amount of nutrients and gamma aminobutyric acid (GABA) were determined. Sprague–Dawley rats were randomly divided into four groups: the control group, FO 50 group (FO 50 mg/kg), and FO 100 group (FO 100 mg/kg) were administered orally once daily and the recombinant human growth hormone (rhGH) group (200 μg/kg) was intraperitoneally injected once daily for 14 days. Results Oral administration of FO 100 significantly increased body length and had no effect on organ damage or hematological profiles. However, administration of rhGH significantly induced hypertrophy of the liver, kidney and spleen along with a marked increase in body length. Tibia length and the growth plate were increased, and bone morphometric parameters were slightly improved by FO and rhGH administration. Serum analysis showed that the levels of GH and insulin like growth factor-1 (IGF-1) were slightly upregulated by FO administration. Nevertheless, the protein expression of hepatic IGF-1 was markedly increased by FO 100 and rhGH administration. Conclusions FO have high content of GABA, and induced positive effects on body length, tibial length, growth-plate length and hepatic IGF-1 synthesis in SD rats with no toxicity or alterations of hematological profile. Therefore, these results suggest that GABA-enriched FO could be considered a potential alternative treatment for growth stimulation.
Collapse
Affiliation(s)
- Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| | - Hyun Hwang-Bo
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| | - Minji Woo
- Busan Innovation Institute of Industry, Science & Technology Planning, Busan, Republic of Korea
| | - Young-Sam Keum
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, Republic of Korea
| | - Jeong Sook Noh
- Department of Food Science & Nutrition, Tongmyong University, Busan, Republic of Korea
| | - Joung-Hyun Park
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan, Republic of Korea
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Eui Kyun Park
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
| |
Collapse
|
38
|
Blagodatski A, Klimenko A, Jia L, Katanaev VL. Small Molecule Wnt Pathway Modulators from Natural Sources: History, State of the Art and Perspectives. Cells 2020; 9:cells9030589. [PMID: 32131438 PMCID: PMC7140537 DOI: 10.3390/cells9030589] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023] Open
Abstract
The Wnt signaling is one of the major pathways known to regulate embryonic development, tissue renewal and regeneration in multicellular organisms. Dysregulations of the pathway are a common cause of several types of cancer and other diseases, such as osteoporosis and rheumatoid arthritis. This makes Wnt signaling an important therapeutic target. Small molecule activators and inhibitors of signaling pathways are important biomedical tools which allow one to harness signaling processes in the organism for therapeutic purposes in affordable and specific ways. Natural products are a well known source of biologically active small molecules with therapeutic potential. In this article, we provide an up-to-date overview of existing small molecule modulators of the Wnt pathway derived from natural products. In the first part of the review, we focus on Wnt pathway activators, which can be used for regenerative therapy in various tissues such as skin, bone, cartilage and the nervous system. The second part describes inhibitors of the pathway, which are desired agents for targeted therapies against different cancers. In each part, we pay specific attention to the mechanisms of action of the natural products, to the models on which they were investigated, and to the potential of different taxa to yield bioactive molecules capable of regulating the Wnt signaling.
Collapse
Affiliation(s)
- Artem Blagodatski
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690090, Russia;
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Correspondence: (A.B.); (V.L.K.)
| | - Antonina Klimenko
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690090, Russia;
| | - Lee Jia
- Institute of Oceanography, Minjiang University, Fuzhou 350108, China;
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Vladimir L. Katanaev
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690090, Russia;
- Institute of Oceanography, Minjiang University, Fuzhou 350108, China;
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: (A.B.); (V.L.K.)
| |
Collapse
|