1
|
Goleij P, Sanaye PM, Alam W, Zhang J, Tabari MAK, Filosa R, Jeandet P, Cheang WS, Efferth T, Khan H. Unlocking daidzein's healing power: Present applications and future possibilities in phytomedicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155949. [PMID: 39217652 DOI: 10.1016/j.phymed.2024.155949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cancer is one of the leading causes of death and a great threat to people around the world. Cancer treatment modalities include surgery, radiotherapy, chemotherapy, radiochemotherapy, hormone therapy, and immunotherapy. The best approach is to use a combination of several types. Among the treatment methods mentioned above, chemotherapy is frequently used, but its activity is hampered by the development of drug resistance and many side effects. In this regard, the use of medicinal plants has been discussed, and in recent decades, the use of isolated phytochemicals came into the focus of interest. By critically evaluating the available evidence and emphasizing the unique perspective offered by this review, we provide insights into the potential of daidzein as a promising therapeutic agent, as well as outline future research directions to optimize its efficacy in clinical settings. PURPOSE To summarized the therapeutic potential of daidzein, an isoflavone phytoestrogen in the management of several human diseases with the focuses on the current status and future prospects as a therapeutic agent. METHODS Several search engines, including PubMed, GoogleScholar, and ScienceDirect, were used, with the search terms "daidzein", "daidzein therapeutic potential", or individual effects. The study included all peer-reviewed articles. However, the most recent publications were given priority. RESULTS Daidzein showed protective effects against malignant diseases such as breast cancer, prostate cancer but also non-malignant diseases such as diabetes, osteoporosis, and cardiovascular diseases. Daidzein activates multiple signaling pathways leading to cell cycle arrest and apoptosis as well as antioxidant and anti-metastatic effects in malignant cells. Moreover, the anticancer effects against different cancer cells were more prominent and discussed in detail. CONCLUSIONS In short, daidzein represents a promising compound for drug development. The comprehensive potential anticancer activities of daidzein through various molecular mechanisms and its therapeutic/clinical status required further detail studies.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran.
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Waqas Alam
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Mohammad Amin Khazeei Tabari
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Rosanna Filosa
- Department of Science and Technology, University of Sannio, Benevento 82100, Italy
| | - Philippe Jeandet
- Département de Biologie et Biochimie Faculté des Sciences Exactes et Naturelles Université de Reims BP 1039 51687, Reims CEDEX 02, France
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| |
Collapse
|
2
|
Zhao Q, Feng J, Liu F, Liang Q, Xie M, Dong J, Zou Y, Ye J, Liu G, Cao Y, Guo Z, Qiao H, Zheng L, Zhao K. Rhizoma Drynariae-derived nanovesicles reverse osteoporosis by potentiating osteogenic differentiation of human bone marrow mesenchymal stem cells via targeting ER α signaling. Acta Pharm Sin B 2024; 14:2210-2227. [PMID: 38799625 PMCID: PMC11119514 DOI: 10.1016/j.apsb.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 05/29/2024] Open
Abstract
Although various anti-osteoporosis drugs are available, the limitations of these therapies, including drug resistance and collateral responses, require the development of novel anti-osteoporosis agents. Rhizoma Drynariae displays a promising anti-osteoporosis effect, while the effective component and mechanism remain unclear. Here, we revealed the therapeutic potential of Rhizoma Drynariae-derived nanovesicles (RDNVs) for postmenopausal osteoporosis and demonstrated that RDNVs potentiated osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) by targeting estrogen receptor-alpha (ERα). RDNVs, a natural product isolated from fresh Rhizoma Drynariae root juice by differential ultracentrifugation, exhibited potent bone tissue-targeting activity and anti-osteoporosis efficacy in an ovariectomized mouse model. RDNVs, effectively internalized by hBMSCs, enhanced proliferation and ERα expression levels of hBMSC, and promoted osteogenic differentiation and bone formation. Mechanistically, via the ERα signaling pathway, RDNVs facilitated mRNA and protein expression of bone morphogenetic protein 2 and runt-related transcription factor 2 in hBMSCs, which are involved in regulating osteogenic differentiation. Further analysis revealed that naringin, existing in RDNVs, was the active component targeting ERα in the osteogenic effect. Taken together, our study identified that naringin in RDNVs displays exciting bone tissue-targeting activity to reverse osteoporosis by promoting hBMSCs proliferation and osteogenic differentiation through estrogen-like effects.
Collapse
Affiliation(s)
- Qing Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Junjie Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fubin Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qianxin Liang
- The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, China
| | - Manlin Xie
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiaming Dong
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanfang Zou
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiali Ye
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guilong Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Blood Transfusion, Guangdong Heyou International Hospital, Foshan 528306, China
| | - Yue Cao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhaodi Guo
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongzhi Qiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Zheng
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kewei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
3
|
Singh S, Verma SC, Kumar V, Sharma K, Singh D, Khan S, Gupta N, Singh R, Khan F, Chanda D, Mishra DP, Singh D, Roy P, Gupta A. Synthesis of amide derivatives of 3-aryl-3H-benzopyrans as osteogenic agent concomitant with anticancer activity. Bioorg Chem 2023; 133:106380. [PMID: 36731295 DOI: 10.1016/j.bioorg.2023.106380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/02/2022] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
The present study reports a series of 3-aryl-3H-benzopyran-based amide derivatives as osteogenic agents concomitant with anticancer activity. Six target compounds viz 22e, 22f, 23i, and 24b-d showed good osteogenic activity at 1 pM and 100 pM concentrations. One of the potential molecules, 24b, effectively induced ALP activity and mRNA expression of osteogenic marker genes at 1 pM and bone mineralization at 100 pM concentrations. These molecules also presented significant growth inhibition of osteosarcoma (MG63) and estrogen-dependent and -independent (MCF-7 and MDA-MB-231) breast cancer cells. The most active compound, 24b, inhibited the growth of all the cancer cells within the IC50 10.45-12.66 µM. The mechanistic studies about 24b showed that 24b induced apoptosis via activation of the Caspase-3 enzyme and inhibited cancer cell migration. In silico molecular docking performed for 24b revealed its interaction with estrogen receptor-β (ER-β) preferentially.
Collapse
Affiliation(s)
- Sarita Singh
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Surendra Chandra Verma
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India
| | - Vinay Kumar
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Kriti Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram extension, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Diksha Singh
- Bioprospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India
| | - Sana Khan
- Technology Dissemination and Computational Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India
| | - Neelam Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram extension, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Romila Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram extension, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Feroz Khan
- Technology Dissemination and Computational Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Debabrata Chanda
- Bioprospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Durga Prasad Mishra
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram extension, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Divya Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram extension, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Atul Gupta
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Kukrail Road, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR),Ghaziabad, Uttar Pradesh- 201002, India.
| |
Collapse
|
4
|
Estrogenic flavonoids and their molecular mechanisms of action. J Nutr Biochem 2023; 114:109250. [PMID: 36509337 DOI: 10.1016/j.jnutbio.2022.109250] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Flavonoids are a major group of phytoestrogens associated with physiological effects, and ecological and social impacts. Although the estrogenic activity of flavonoids was reported by researchers in the fields of medical, environmental and food studies, their molecular mechanisms of action have not been comprehensively reviewed. The estrogenic activity of the respective classes of flavonoids, anthocyanidins/anthocyanins, 2-arylbenzofurans/3-arylcoumarins/α-methyldeoxybenzoins, aurones/chalcones/dihydrochalcones, coumaronochromones, coumestans, flavans/flavan-3-ols/flavan-4-ols, flavanones/dihydroflavonols, flavones/flavonols, homoisoflavonoids, isoflavans, isoflavanones, isoflavenes, isoflavones, neoflavonoids, oligoflavonoids, pterocarpans/pterocarpenes, and rotenone/rotenoids, was summarized through a comprehensive literature search, and their structure-activity relationship, biological activities, signaling pathways, and applications were discussed. Although the respective classes of flavonoids contained at least one chemical mimicking estrogen, the mechanisms varied, such as those with estrogenic, anti-estrogenic, non-estrogenic, and biphasic activities, and additional activities through crosstalk/bypassing, which exert biological activities through cell signaling pathways. Such mechanistic variations of estrogen action are not limited to flavonoids and are observed among other broad categories of chemicals, thus this group of chemicals can be termed as the "estrogenome". This review article focuses on the connection of estrogen action mainly between the outer and the inner environments, which represent variations of chemicals and biological activities/signaling pathways, respectively, and form the basis to understand their applications. The applications of chemicals will markedly progress due to emerging technologies, such as artificial intelligence for precision medicine, which is also true of the study of the estrogenome including estrogenic flavonoids.
Collapse
|
5
|
Li H, Wang C, Jin Y, Cai Y, Yao J, Meng Q, Wu J, Wang H, Sun H, Liu M. Anti-Postmenopausal osteoporosis effects of Isopsoralen: A bioinformatics-integrated experimental study. Phytother Res 2023; 37:231-251. [PMID: 36123318 DOI: 10.1002/ptr.7609] [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: 05/08/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Abstract
Isopsoralen (IPRN), which comes from the fruit of Psoralea corylifolia, has been identified as a kind of phytoestrogen and has been proven to be effective for the treatment of osteoporosis (OP). However, the mechanisms underlying IPRN's anti-OP effects, especially the anti-postmenopausal osteoporosis (PMOP) effects, remain indistinct. Thus, this study aimed to investigate the effects and mechanisms of IPRN's anti-PMOP activity. In this study, the bioinformatics results predicted that IPRN could resist PMOP by targeting EGFR, AKT1, SRC, CCND1, ESR1 (ER-α), AR, PGR, BRCA1, PTGS2, and IGF1R. An ovariectomized (OVX) mice model and a H2 O2 -induced bone marrow mesenchyml stem cells (BMSCs) model confirmed that IPRN could inhibit the bone loss induced by OVX in mice and promote the osteogenic differentiation in H2 O2 -induced BMSCs by inhibiting oxidative stress and apoptosis. Moreover, IPRN could significantly produce the above effects by upregulating ESR1. IPRN might be a therapeutic agent for PMOP by acting as an estrogen replacement agent and a natural antioxidant.
Collapse
Affiliation(s)
- Hao Li
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yue Jin
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yuanqing Cai
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jialin Yao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huihan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Foley JP, Fred EJ, Minardi S, Yamaguchi JT, Greene AC, Furman AA, Lyons JG, Paul JT, Nandurkar TS, Blank KR, Havey RM, Muriuki M, Patwardhan AG, Hsu WK, Stock SR, Hsu EL. Sex-based Difference in Response to Recombinant Human Bone Morphogenetic Protein-2 in a Rat Posterolateral Fusion Model. Spine (Phila Pa 1976) 2022; 47:1627-1636. [PMID: 35943241 PMCID: PMC9643612 DOI: 10.1097/brs.0000000000004454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN This was a preclinical study. OBJECTIVE Evaluate sex-dependent differences in the bone healing response to recombinant human bone morphogenetic protein-2 (rhBMP-2) in a rat posterolateral spinal fusion model. SUMMARY OF BACKGROUND DATA Minimal and conflicting data exist concerning potential sex-dependent differences in rhBMP-2-mediated bone regeneration in the context of spinal fusion. MATERIALS AND METHODS Forty-eight female and male Sprague-Dawley rats (N=24/group), underwent L4-L5 posterolateral fusion with bilateral placement of an absorbable collagen sponge, each loaded with 5 µg of bone morphogenetic protein-2 (10 µg/animal). At eight weeks postoperative, 10 specimens of each sex were tested in flexion-extension with quantification of range of motion and stiffness. The remaining specimens were evaluated for new bone growth and successful fusion via radiography, blinded manual palpation and microcomputed tomography (microCT). Laboratory microCT quantified bone microarchitecture, and synchrotron microCT examined bone microstructure at the 1 µm level. RESULTS Manual palpation scores differed significantly between sexes, with mean fusion scores of 2.4±0.4 in females versus 3.1±0.6 in males ( P <0.001). Biomechanical stiffness did not differ between sexes, but range of motion was significantly greater and more variable for females versus males (3.7±5.6° vs. 0.27±0.15°, P <0.005, respectively). Laboratory microCT showed significantly smaller volumes of fusion masses in females versus males (262±87 vs. 732±238 mm 3 , respectively, P <0.001) but significantly higher bone volume fraction (0.27±0.08 vs. 0.12±0.05, respectively, P <0.001). Mean trabecular thickness was not different, but trabecular number was significantly greater in females (3.1±0.5 vs. 1.5±0.4 mm -1 , respectively, P <0.001). Synchrotron microCT showed fine bone structures developing in both sexes at the eight-week time point. CONCLUSIONS This study demonstrates sex-dependent differences in bone regeneration induced by rhBMP-2. Further investigation is needed to uncover the extent of and mechanisms underlying these sex differences, particularly at different doses of rhBMP-2.
Collapse
Affiliation(s)
- James P Foley
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Elianna J Fred
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Silvia Minardi
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Jonathan T Yamaguchi
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Allison C Greene
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Andrew A Furman
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Joseph G Lyons
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Jonathan T Paul
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Tejas S Nandurkar
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | | | - Robert M Havey
- Edward Hines Jr. VA Hospital, Hines, IL
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Chicago, Maywood, IL
| | | | - Avinash G Patwardhan
- Edward Hines Jr. VA Hospital, Hines, IL
- Department of Orthopaedic Surgery and Rehabilitation, Loyola University Chicago, Maywood, IL
| | - Wellington K Hsu
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| | - Stuart R Stock
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
- Department of Cell & Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Erin L Hsu
- Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Center for Regenerative Nanomedicine, Simpson Querrey Institute, Northwestern University, Chicago, IL
| |
Collapse
|
7
|
Knewtson KE, Ohl NR, Robinson JL. Estrogen Signaling Dictates Musculoskeletal Stem Cell Behavior: Sex Differences in Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:789-812. [PMID: 34409868 PMCID: PMC9419932 DOI: 10.1089/ten.teb.2021.0094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sexual dimorphisms in humans and other species exist in visually evident features such as body size and less apparent characteristics, including disease prevalence. Current research is adding to a growing understanding of sex differences in stem cell function and response to external stimuli, including sex hormones such as estrogens. These differences are proving significant and directly impact both the understanding of stem cell processes in tissue repair and the clinical implementation of stem cell therapies. Adult stem cells of the musculoskeletal system, including those used for development and repair of muscle, bone, cartilage, fibrocartilage, ligaments, and tendons, are no exception. Both in vitro and in vivo studies have found differences in stem cell number, proliferative and differentiation capabilities, and response to estrogen treatment between males and females of many species. Maintaining the stemness and reducing senescence of adult stem cells is an important topic with implications in regenerative therapy and aging. As such, this review discusses the effect of estrogens on musculoskeletal system stem cell response in multiple species and highlights the research gaps that still need to be addressed. The following evidence from investigations of sex-related phenotypes in adult progenitor and stem cells are pieces to the big puzzle of sex-related effects on aging and disease and critical information for both fundamental tissue repair and regeneration studies and safe and effective clinical use of stem cells. Impact Statement This review summarizes current knowledge of sex differences in and the effects of estrogen treatment on musculoskeletal stem cells in the context of tissue engineering. Specifically, it highlights the impact of sex on musculoskeletal stem cell function and ability to regenerate tissue. Furthermore, it discusses the varying effects of estrogen on stem cell properties, including proliferation and differentiation, important to tissue engineering. This review aims to highlight the potential impact of estrogens and the importance of performing sex comparative studies in the field of tissue engineering.
Collapse
Affiliation(s)
- Kelsey E. Knewtson
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Nathan R. Ohl
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Jennifer L. Robinson
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Address correspondence to: Jennifer L. Robinson, PhD, Department of Chemical and Petroleum Engineering, The University of Kansas, 1530 West 15th Street Room 4132, Lawrence, KS 66045, USA
| |
Collapse
|
8
|
Li J, Xu F, Ji D, Tian C, Sun Y, Mutanda I, Ren Y, Wang Y. Diversion of metabolic flux towards 5-deoxy(iso)flavonoid production via enzyme self-assembly in Escherichia coli. Metab Eng Commun 2021; 13:e00185. [PMID: 34631421 PMCID: PMC8488244 DOI: 10.1016/j.mec.2021.e00185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022] Open
Abstract
5-Deoxy(iso)flavonoids are structural representatives of phenylpropanoid-derived compounds and play critical roles in plant ecophysiology. Recently, 5-deoxy(iso)flavonoids gained significant interest due to their potential applications as pharmaceuticals, nutraceuticals, and food additives. Given the difficulties in their isolation from native plant sources, engineered biosynthesis of 5-deoxy(iso)flavonoids in a microbial host is a highly promising alternative approach. However, the production of 5-deoxy(iso)flavonoids is hindered by metabolic flux imbalances that result in a product profile predominated by non-reduced analogues. In this study, GmCHS7 (chalcone synthase from Glycine max) and GuCHR (chalcone reductase from Glycyrrhizza uralensis) were preliminarily utilized to improve the CHR ratio (CHR product to total CHS product). The use of this enzyme combination improved the final CHR ratio from 39.7% to 50.3%. For further optimization, a protein-protein interaction strategy was employed, basing on the spatial adhesion of GmCHS7:PDZ and GuCHR:PDZlig. This strategy further increased the ratio towards the CHR-derived product (54.7%), suggesting partial success of redirecting metabolic flux towards the reduced branch. To further increase the total carbon metabolic flux, 15 protein scaffolds were programmed with stoichiometric arrangement of the three sequential catalysts GmCHS7, GuCHR and MsCHI (chalcone isomerase from Medicago sativa), resulting in a 1.4-fold increase in total flavanone production, from 69.4 mg/L to 97.0 mg/L in shake flasks. The protein self-assembly strategy also improved the production and direction of the lineage-specific compounds 7,4'-dihydroxyflavone and daidzein in Escherichia coli. This study presents a significant advancement of 5-deoxy(iso)flavonoid production and provides the foundation for production of value-added 5-deoxy(iso)flavonoids in microbial hosts.
Collapse
Affiliation(s)
- Jianhua Li
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Fanglin Xu
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- He'nan Key Laboratory of Plant Stress Biology, He'nan University, Kaifeng, 475004, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Dongni Ji
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Chenfei Tian
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yuwei Sun
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Ishmael Mutanda
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yuhong Ren
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yong Wang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
9
|
Ren W, Gan D, Tan G, Xue H, Li N, Xu Z. CHANGES OF WNT/B-CATENIN SIGNALING AND DIFFERENTIATION POTENTIAL OF BONE MARROW MESENCHYMAL STEM CELLS IN PROCESS OF BONE LOSS IN OVARIECTOMIZED RATS. ACTA ENDOCRINOLOGICA-BUCHAREST 2020; 16:156-164. [PMID: 33029231 DOI: 10.4183/aeb.2020.156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background In vitro studies of the changes about osteoblastogenesis and adipogenesis potential of BMSCs were not clear. As it is the critical pathway for osteogenic differentiation and bone formation, whether or not Wnt/β-catenin signalling is involved in the changes of osteogenic and adipogenic potential of BMSCs and participates in bone content decrease of ovariectomized (OVX)osteoporosis rats has been rarely reported. Material/Methods BMSCs from femurs of ovariectomzed rats were isolated and cultured in vitro. The proliferation potential of BMSCs was analysed by CCK-8 assays . Osteoblastic and adipogenic differentiation potential of the BMSCs was assessed by ALP activity assay, Alizarin red S staining, Oil red O staining and RT-PCR analysis. Results The results demonstrated that BMSCs from bilateral ovariectomization rats were endowed with lower proliferation and osteoblastic differentiation potential but higher adipogenic potential than the control group in vitro. In addition, β-catenin was found to have been decreased in OVX BMSCs, indicating that Wnt/β-catenin signalling pathways were suppressed in OVX BMSCs . Conclusions Results suggested that changes in the Wnt canonical signalling pathway may be related to imbalances of osteogenic and adipogenic potential of BMSCs, and this may be an important factor related to bone content decrease in ovariectomized osteoporosis rats.
Collapse
|
10
|
Zhou S, Huang G, Chen G. Synthesis and biological activities of drugs for the treatment of osteoporosis. Eur J Med Chem 2020; 197:112313. [PMID: 32335412 DOI: 10.1016/j.ejmech.2020.112313] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
Osteoporosis is an asymptomatic progressive disease. With the improvement of people's living standard and the aging of population, osteoporosis and its fracture have become one of the main diseases threatening the aging society. The serious medical and social burden caused by this has aroused wide public concern. Osteoporosis is listed as one of the three major diseases of the elderly. At present, the drugs for osteoporosis include bone resorption inhibitors and bone formation promoters. The purpose of these anti-osteoporosis drugs is to balance osteoblast bone formation and osteoclast bone resorption. With the development of anti-osteoporosis drugs, new anti osteoporosis drugs have been designed and synthesized. There are many kinds of new compounds with anti osteoporosis activity, but most of them are concentrated on the original drugs with anti osteoporosis activity, or the natural products with anti-osteoporosis activity are extracted from the natural products for structural modification to obtain the corresponding derivatives or analogues. These target compounds showed good ALP activity in vitro and in vivo, promoted osteoblast differentiation and mineralization, or had anti TRAP activity, inhibited osteoclast absorption. This work attempts to systematically review the studies on the synthesis and bioactivity of anti-osteoporosis drugs in the past 10 years. The structure-activity relationship was discussed, which provided a reasonable idea for the design and development of new anti-osteoporosis drugs.
Collapse
Affiliation(s)
- Shiyang Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Gangliang Huang
- Active Carbohydrate Research Institute, Chongqing Key Laboratory of Green Synthesis and Application, College of Chemistry, Chongqing Normal University, Chongqing, 401331, China.
| | - Guangying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China.
| |
Collapse
|
11
|
Liu ZM, Chen B, Li S, Li G, Zhang D, Ho SC, Chen YM, Ma J, Qi H, Ling WH. Effect of whole soy and isoflavones daidzein on bone turnover and inflammatory markers: a 6-month double-blind, randomized controlled trial in Chinese postmenopausal women who are equol producers. Ther Adv Endocrinol Metab 2020; 11:2042018820920555. [PMID: 32595918 PMCID: PMC7303504 DOI: 10.1177/2042018820920555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/13/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Human studies have demonstrated the beneficial effects of soy or isoflavones on bone metabolism. However, conflicting data remain. Equol is the intestinal metabolite of the isoflavone daidzein. The health benefits of soy are more pronounced in equol producers than those not producing equol. This 6-month randomized controlled trial aimed to examine the effect of whole soy (soy flour) and purified daidzein on bone turnover markers (BTMs) in Chinese postmenopausal women who are equol producers. METHODS A total of 270 eligible women were randomized to either one of the three isocaloric supplements as follows: 40 g soy flour (whole soy group), 40 g low-fat milk powder + 63 mg daidzein (daidzein group), or 40 g low-fat milk powder (placebo group) given as a solid beverage daily for 6 months. The following fasting venous samples were collected at the baseline and end of the trial to analyze BTMs: serum cross-linked C-telopeptides of type I collagen, bone-specific alkaline phosphatase, osteocalcin, procollagen type I N-terminal propeptide, and 25(OH)D3. Inflammation-related biomarkers, such as serum interleukin-6, tumor necrosis factor-alpha, C-reactive protein, transferrin, and homocysteine, were also tested to explore potential mechanisms. RESULTS A total of 253 subjects validly completed the study protocol. Urinary isoflavones suggested a good compliance to the treatments. Intention-to-treat and per-protocol analyses indicated no significant difference in the 6-month or percentage changes in the parameters of bone metabolism and inflammatory markers among the three treatment groups. CONCLUSIONS Whole soy and purified daidzein at provided dosages exhibited no significant effect on the bone metabolism and inflammation levels among Chinese equol-producing postmenopausal women. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT01270737.
Collapse
Affiliation(s)
| | - Bailing Chen
- Department of Spine Surgery, the First
Affiliated Hospital of Sun Yet-sen University, Guangzhou, Guangdong, PR
China
| | - Shuyi Li
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Guoyi Li
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Di Zhang
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Suzanne C. Ho
- Department of Epidemiology, Jockey Club School
of Public Health and Primary Care, the Chinese University of Hong Kong, New
Territories, Hong Kong
| | - Yu-ming Chen
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Jing Ma
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Huang Qi
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| | - Wen-hua Ling
- Department of Nutrition, School of Public
Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, PR
China
| |
Collapse
|
12
|
Peng XY, Hu M, Liao F, Yang F, Ke QF, Guo YP, Zhu ZH. La-Doped mesoporous calcium silicate/chitosan scaffolds for bone tissue engineering. Biomater Sci 2019; 7:1565-1573. [PMID: 30688345 DOI: 10.1039/c8bm01498a] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Trace rare earth elements such as lanthanum (La) regulated effectively bone tissue performances; however, the underlying mechanism remains unknown. In order to accelerate bone defects especially in patients with osteoporosis or other metabolic diseases, we firstly constructed lanthanum-doped mesoporous calcium silicate/chitosan (La-MCS/CTS) scaffolds by freeze-drying technology. During the freeze-drying procedure, three-dimensional macropores were produced within the La-MCS/CTS scaffolds by using ice crystals as templates, and the La-MCS nanoparticles were distributed on the macropore walls. The hierarchically porous structures and biocompatible components contributed to the adhesion, spreading and proliferation of rat bone marrow-derived mesenchymal stem cells (rBMSCs), and accelerated the in-growth of new bone tissues. Particularly, the La3+ ions in the bone scaffolds remarkably induced the osteogenic differentiation of rBMSCs via the activation of the TGF signal pathway. A critical-sized calvarial-defect rat model further revealed that the La-MCS/CTS scaffolds significantly promoted new bone regeneration as compared with pure MCS/CTS scaffolds. In conclusion, the La-MCS/CTS scaffold showed the prominent ability in osteogenesis and bone regeneration, which showed its application potential for bone defect therapy.
Collapse
Affiliation(s)
- Xiao-Yuan Peng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
13
|
Fan H, Lv Z, Gan L, Ning C, Li Z, Yang M, Zhang B, Song B, Li G, Tang D, Gao J, Yan S, Wang Y, Liu J, Guo Y. A Novel lncRNA Regulates the Toll-Like Receptor Signaling Pathway and Related Immune Function by Stabilizing FOS mRNA as a Competitive Endogenous RNA. Front Immunol 2019; 10:838. [PMID: 31057556 PMCID: PMC6478817 DOI: 10.3389/fimmu.2019.00838] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 04/01/2019] [Indexed: 01/01/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have recently emerged as new regulatory molecules with diverse functions in regulating gene expression and significant roles in the immune response. However, the function of many unknown lncRNAs is still unclear. By studying the regulatory effect of daidzein (DA) on immunity, we identified a novel lncRNA with an immune regulatory function: lncRNA- XLOC_098131. In vivo, DA treatment upregulated the expression of lncRNA- XLOC_098131, FOS, and JUN in chickens and affected the expression of activator protein 1 (AP-1) to regulate MAPK signaling, Toll-like receptor signaling, and related mRNA expression. It also enhanced macrophage activity and increased the numbers of blood neutrophils and mononuclear cells, which can improve the body's ability to respond to stress and bacterial and viral infections. Furthermore, DA treatment also reduced B lymphocyte apoptosis and promoted the differentiation of B lymphocytes into plasma cells, which in turn resulted in the production of more immunoglobulins and the promotion of antigen presentation. In vitro, using HEK293FT cells, we demonstrated that mir-548s could bind to and decrease the expression of both FOS and lncRNA- XLOC_098131. LncRNA- XLOC_098131 served as a competitive endogenous RNA to stabilize FOS by competitively binding to miR-548s and thereby reducing its inhibitory effect of FOS expression. Therefore, we concluded that the novel lncRNA XLOC_098131 acts as a key regulatory molecule that can regulate the Toll-like receptor signaling pathway and related immune function by serving as a competitive endogenous RNA to stabilize FOS mRNA expression.
Collapse
Affiliation(s)
- Hao Fan
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zengpeng Lv
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Liping Gan
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chao Ning
- Key Laboratory of Animal Genetics, Breeding and Reproduction, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Ministry of Agriculture, Beijing, China
| | - Zhui Li
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Minghui Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Ministry of Agriculture, Beijing, China
| | - Beibei Zhang
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Bochen Song
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Guang Li
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dazhi Tang
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinxin Gao
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shaojia Yan
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Youli Wang
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianfeng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Ministry of Agriculture, Beijing, China
| | - Yuming Guo
- State key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Comparison among Activities and Isoflavonoids from Pueraria thunbergiana Aerial Parts and Root. Molecules 2019; 24:molecules24050912. [PMID: 30841642 PMCID: PMC6429325 DOI: 10.3390/molecules24050912] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 11/17/2022] Open
Abstract
Kudzu (Pueraria thunbergiana Benth.) has long been used as a food and medicine for many centuries. The root is the most commonly used portion of the plant, but the aerial parts are occasionally used as well. In this study, we investigated the constituent compounds and biological activities of the aerial parts, leaves, stems, and sprouts, and compared their constituents and activities with those of roots. Leaf extract showed a significantly higher TPC level at 59 ± 1.6 mg/g and lower free radical scavenging (FRS) values under 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2’-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid (ABTS), and NO inhibition at 437 ± 11, 121 ± 6.6 μg/mL and 107 ± 4.9 μg/mL, respectively, than those of sprout, stem, and root extract. Leaf extract also significantly suppressed lipopolysaccharide (LPS)-mediated gene expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). The main components of leaf extract were found to be genistin and daidzin. This study suggests that the leaves of kudzu are a good source of biological activities and isoflavones that can be used in functional or medicinal foods and cosmetics for the prevention or treatment of diseases related to inflammation and oxidative stress.
Collapse
|
15
|
Bateman ME, Strong AL, Hunter RS, Bratton MR, Komati R, Sridhar J, Riley KE, Wang G, Hayes DJ, Boue SM, Burow ME, Bunnell BA. Osteoinductive effects of glyceollins on adult mesenchymal stromal/stem cells from adipose tissue and bone marrow. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 27:39-51. [PMID: 28314478 DOI: 10.1016/j.phymed.2017.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/04/2017] [Accepted: 02/12/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND While current therapies for osteoporosis focus on reducing bone resorption, the development of therapies to regenerate bone may also be beneficial. Promising anabolic therapy candidates include phytoestrogens, such as daidzein, which effectively induce osteogenesis of adipose-derived stromal cells (ASCs) and bone marrow stromal cells (BMSCs). PURPOSE To investigate the effects of glyceollins, structural derivatives of daidzein, on osteogenesis of ASCs and BMSCs. STUDY DESIGN Herein, the osteoinductive effects of glyceollin I and glyceollin II were assessed and compared to estradiol in ASCs and BMSCs. The mechanism by which glyceollin II induces osteogenesis was further examined. METHODS The ability of glyceollins to promote osteogenesis of ASCs and BMSCs was evaluated in adherent and scaffold cultures. Relative deposition of calcium was analyzed using Alizarin Red staining, Bichinchoninic acid Protein Assay, and Alamar Blue Assay. To further explore the mechanism by which glyceollin II exerts its osteoinductive effects, docking studies of glyceollin II, RNA isolation, cDNA synthesis, and quantitative RT-PCR (qPCR) were performed. RESULTS In adherent cultures, ASCs and BMSCs treated with estradiol, glyceollin I, or glyceollin II demonstrated increased calcium deposition relative to vehicle-treated cells. During evaluation on PLGA scaffolds seeded with ASCs and BMSCs, glyceollin II was the most efficacious in inducing ASC and BMSC osteogenesis compared to estradiol and glyceollin I. Dose-response analysis in ASCs and BMSCs revealed that glyceollin II has the highest potency at 10nM in adherent cultures and 1µM in tissue scaffold cultures. At all doses, osteoinductive effects were attenuated by fulvestrant, suggesting that glyceollin II acts at least in part through estrogen receptor-mediated pathways to induce osteogenesis. Analysis of gene expression demonstrated that, similar to estradiol, glyceollin II induces upregulation of genes involved in osteogenic differentiation. CONCLUSION The ability of glyceollin II to induce osteogenic differentiation in ASCs and BMSCs indicates that glyceollins hold the potential for the development of pharmacological interventions to improve clinical outcomes of patients with osteoporosis.
Collapse
Affiliation(s)
- Marjorie E Bateman
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ryan S Hunter
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Melyssa R Bratton
- Cell and Molecular Biology Core Facility, Xavier University of Louisiana, New Orleans, LA, USA
| | - Rajesh Komati
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Jayalakshmi Sridhar
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Kevin E Riley
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Daniel J Hayes
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, USA
| | - Stephen M Boue
- Southern Regional Research Center, US Department of Agriculture, 1100 Robert E. Lee Blvd, New Orleans, LA, USA
| | - Matthew E Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA; Division of Regenerative Medicine, Tulane National Primate Research Center, Tulane University, Covington, LA USA.
| |
Collapse
|
16
|
Kumagai M, Nishikawa K, Mishima T, Yoshida I, Ide M, Koizumi K, Nakamura M, Morimoto Y. Synthesis of novel 5,6-dehydrokawain analogs as osteogenic inducers and their action mechanisms. Bioorg Med Chem Lett 2017; 27:2401-2406. [PMID: 28427810 DOI: 10.1016/j.bmcl.2017.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022]
Abstract
An imbalance between bone resorption by osteoclasts and bone formation by osteoblasts can cause bone loss and bone-related disease. In a previous search for natural products that increase osteogenic activity, we found that 5,6-dehydrokawain (1) from Alpinia zerumbet promotes osteoblastogenesis. In this study, we synthesized and evaluated series of 5,6-dehydrokawain analogs. Our structure-activity relationships revealed that alkylation of para or meta position of aromatic ring of 1 promote osteogenic activity. Among the potential analogs we synthesized, (E)-6-(4-Ethylstyryl)-4-methoxy-2H-pyran-2-one (14) and (E)-6-(4-Butylstyryl)-4-methoxy-2H-pyran-2-one (21) both significantly up-regulated Runx2 and Osterix mRNA expression at 10µM. These osteogenic activities could be mediated by bone morphogenetic protein (BMP) and activation of p38 MAPK signaling pathways. Compounds 14 and 21 also inhibited RANKL-induced osteoclast differentiation of RAW264 cells. These results indicated that novel 5,6-dehydrokawain analogs not only increase osteogenic activity but also inhibit osteoclast differentiation, and could be potential lead compounds for the development of anti-osteoporosis agents.
Collapse
Affiliation(s)
- Momochika Kumagai
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan; Department of Chemistry, Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka 558-8585, Japan.
| | - Keisuke Nishikawa
- Department of Chemistry, Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Takashi Mishima
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan
| | - Izumi Yoshida
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan
| | - Masahiro Ide
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan
| | - Keiko Koizumi
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan
| | - Munetomo Nakamura
- Department of Research and Development, Japan Food Research Laboratories, Osaka 567-0085, Japan
| | - Yoshiki Morimoto
- Department of Chemistry, Graduate School of Science, Osaka City University, Sumiyoshi-ku, Osaka 558-8585, Japan
| |
Collapse
|
17
|
Mollazadeh S, Neshati V, Fazly Bazzaz BS, Iranshahi M, Mojarrad M, Naderi-Meshkin H, Kerachian MA. StandardizedSophora pachycarpaRoot Extract Enhances Osteogenic Differentiation in Adipose-derived Human Mesenchymal Stem Cells. Phytother Res 2017; 31:792-800. [DOI: 10.1002/ptr.5803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/20/2017] [Accepted: 02/24/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Samaneh Mollazadeh
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Vajiheh Neshati
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Genetics, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group; Academic Center for Education, Culture and Research (ACECR), Khorasan-Razavi Branch; Mashhad Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Genetics, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
18
|
Murugan Girija D, Y Ranga Rao S, Kalachaveedu M, Subbarayan R. Osteogenic differentiation of human gingival mesenchymal stem cells byAristolochia bracteolatasupplementation through enhanced Runx2 expression. J Cell Physiol 2017; 232:1591-1595. [DOI: 10.1002/jcp.25835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Dinesh Murugan Girija
- Centre for Indian Systems of Medicine Quality Assurance and Standardization Sri Ramachandra University; Chennai India
| | | | | | - Rajasekaran Subbarayan
- Centre for Regenerative Medicine and Stem Cell Research; Sri Ramachandra University; Chennai India
| |
Collapse
|
19
|
Gupta A, Ahmad I, Kureel J, John AA, Sultan E, Chanda D, Agarwal NK, Alauddin, Wahajuddin, Prabhaker S, Verma A, Singh D. Differentiation of skeletal osteogenic progenitor cells to osteoblasts with 3,4-diarylbenzopyran based amide derivatives: Novel osteogenic agents. Eur J Med Chem 2016; 121:82-99. [DOI: 10.1016/j.ejmech.2016.05.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/08/2016] [Accepted: 05/07/2016] [Indexed: 01/24/2023]
|
20
|
17β-estradiol differently affects osteogenic differentiation of mesenchymal stem/stromal cells from adipose tissue and bone marrow. Differentiation 2016; 92:291-297. [PMID: 27087652 DOI: 10.1016/j.diff.2016.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/14/2016] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
Abstract
Adipose-derived and bone marrow stem/stromal cells (ASCs and BMSCs) have been often compared for their application in regenerative medicine, and several factors sustaining their differentiation and efficacy have been investigated. 17 β-estradiol (E2) has been reported to influence some functions of progenitor cells. Here we studied the effects of 10 and 100nM E2 on ASC and BMSC vitality, proliferation and differentiation towards osteogenic and adipogenic lineages. E2 did not modulate ASC and BMSC vitality and growth rate, while the hormone produced a pro-adipogenic effect on both mesenchymal stem/stromal cells (MSCs). In particular, the synergy between 7-day pre-treatment and 100nM E2 led to the most evident result, increasing lipid vacuoles formation in ASCs and BMSCs of +44% and +82%, respectively. Despite the fact that E2 did not alter collagen deposition of osteo-induced MSCs, we observed a different modulation of ASC and BMSC alkaline phosphatase (ALP) activity. Indeed, this osteogenic marker was always enhanced by 17 β-estradiol in BMSCs, and 7-day pre-treatment with 100nM E2 increased it of about 70%. In contrast, E2 weakened ASC osteogenic potential, reducing their ALP activity of about 20%, with the most evident effect on ASCs isolated from pre-menopausal women (-30%). Finally, we identified an estrogen receptor α (ERα) variant of about 37kDa expressed in both MSCs. Interestingly, adipogenic stimuli drastically reduced its expression, while osteogenic ones mildly increased this isoform in BMSCs only. In conclusion, E2 positively affected the adipogenic process of both MSCs while it favored osteogenic induction in BMSCs only, and both mesenchymal progenitors expressed a novel 37kDa ER-α variant whose expression was modulated during differentiation.
Collapse
|
21
|
Liu H, Li W, Liu Y, Zhang X, Zhou Y. Co-administration of aspirin and allogeneic adipose-derived stromal cells attenuates bone loss in ovariectomized rats through the anti-inflammatory and chemotactic abilities of aspirin. Stem Cell Res Ther 2015; 6:200. [PMID: 26474767 PMCID: PMC4609080 DOI: 10.1186/s13287-015-0195-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/20/2015] [Accepted: 10/01/2015] [Indexed: 01/21/2023] Open
Abstract
Introduction Osteoporosis is a syndrome of excessive skeletal fragility characterized by the loss of mass and deterioration of microarchitecture in bone. Single use of aspirin or adipose-derived stromal cells (ASCs) has been recognized recently to be effective against osteoporosis. The goal of the study was to evaluate the osteogenic effects of the co-administration of aspirin and allogeneic rat adipose-derived stromal cells (rASCs) on ovariectomized (OVX)-induced bone loss in rats. The underlying mechanisms were investigated in vitro and in vivo. Methods Firstly, allogeneic rASCs were isolated and cultured, and the conditioned medium (CM) from the maintenance of rASCs was collected. Secondly, the OVX rats were administrated CM, rASCs, aspirin (ASP) or rASCs + ASP, respectively. Twelve weeks later, the anti-inflammatory and osteogenic effects were assessed by micro-CT, undecalcified histological sections, dynamic histomorphometric analyses and serologic assays for biochemical markers. Finally, a Transwell migration assay in vitro and cell-trafficking analyses in vivo were used to explore the effects of aspirin on rASC migration. Results Systemic administration of aspirin and rASCs attenuated OVX-induced bone loss better than single use of aspirin or ASCs (p < 0.05, respectively). Next, we analyzed the underlying mechanisms of the anti-inflammatory and chemotactic abilities of aspirin. Aspirin suppressed serum levels of the pro-inflammatory cytokines on tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and the anti-inflammatory ability was positively associated with bone morphometry. Also, aspirin exhibited excellent chemotactic effects in vitro and accelerated the homing of allogeneic rASCs into bone marrow during early in vivo stages. Conclusions Co-administered aspirin and allogeneic ASCs can partially reverse OVX-induced bone loss in rats. This effect appears to be mediated by the anti-inflammatory and chemotactic abilities of aspirin.
Collapse
Affiliation(s)
- Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Wei Li
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China. .,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
22
|
Sung JH, An HS, Jeong JH, Shin S, Song SY. Megestrol Acetate Increases the Proliferation, Migration, and Adipogenic Differentiation of Adipose-Derived Stem Cells via Glucocorticoid Receptor. Stem Cells Transl Med 2015; 4:789-99. [PMID: 25972147 DOI: 10.5966/sctm.2015-0009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/08/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : Because adipose-derived stem cells (ASCs) are usually expanded to acquire large numbers of cells for therapeutic applications, it is important to increase the production yield and regenerative potential during expansion. Therefore, a tremendous need exists for alternative ASC stimuli during cultivation to increase the proliferation and adipogenic differentiation of ASCs. The present study primarily investigated the involvement of megestrol acetate (MA), a progesterone analog, in the stimulation of ASCs, and identifies the target receptors underlying stimulation. Mitogenic and adipogenic effects of MA were investigated in vitro, and pharmacological inhibition and small interfering (si) RNA techniques were used to identify the molecular mechanisms involved in the MA-induced stimulation of ASCs. MA significantly increased the proliferation, migration, and adipogenic differentiation of ASCs in a dose-dependent manner. Glucocorticoid receptor (GR) is highly expressed compared with other nuclear receptors in ASCs, and this receptor is phosphorylated after MA treatment. MA also upregulated genes downstream of GR in ASCs, including ANGPTL4, DUSP1, ERRF11, FKBP5, GLUL, and TSC22D3. RU486, a pharmacological inhibitor of GR, and transfection of siGR significantly attenuated MA-induced proliferation, migration, and adipogenic differentiation of ASCs. Although the adipogenic differentiation potential of MA was inferior to that of dexamethasone, MA had mitogenic effects in ASCs. Collectively, these results indicate that MA increases the proliferation, migration, and adipogenic differentiation of ASCs via GR phosphorylation. SIGNIFICANCE Magestrol acetate (MA) increases the proliferation, migration, and adipogenic differentiation of adipose-derived stem cells (ASCs) via glucocorticoid receptor phosphorylation. Therefore, MA can be applied to increase the production yield during expansion and can be used to facilitate adipogenic differentiation of ASCs.
Collapse
Affiliation(s)
- Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo-Sun An
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Hyun Jeong
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soyoung Shin
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Yong Song
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|